1
|
Xie Z, Jin L, Wu Q, Ji H, Miao M, Song X, Zhu H, Su H, Yuan W, Liang H. Maternal per- and polyfluoroalkyl substance concentrations and placental DNA methylation of thyroid hormone-related genes. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137545. [PMID: 39947079 DOI: 10.1016/j.jhazmat.2025.137545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 01/10/2025] [Accepted: 02/07/2025] [Indexed: 04/16/2025]
Abstract
Studies suggested that per- and polyfluoroalkyl substances (PFAS) may have adverse effects on fetus by altering placental DNA methylation. In the study, we explored associations of maternal PFAS concentrations with placental DNA methylation of thyroid hormone (TH)-related genes and the potential mediating role of DNA methylation levels in PFAS-fetal TH associations. We measured PFAS concentrations in maternal plasma in early pregnancy and levels of total triiodothyronine (TT3), total thyroxine (TT4), free triiodothyronine (FT3), free thyroxine (FT4), and thyroid stimulating hormone (TSH) in cord plasma. We assessed DNA methylation in 345 placental samples for five TH-related genes, i.e., iodothyronine deiodinase 3 (DIO3), solute carrier family 16 member 2 (SLC16A2), solute carrier organic anion transporter family member 1C1 (SLCO1C1), thyrotropin-releasing hormone (TRH), and transthyretin (TTR). We found the associations of PFDA with increased SLC16A2 methylation, PFOA with decreased SLCO1C1 methylation, PFOS with increased TRH methylation, and PFDoA with decreased TRH methylation, and these associations showed sex-specific patterns. Mediation analyses suggested placental SLCO1C1 and SLC16A2 methylation as potential mediators in the associations of PFOA with FT3 and PFUdA with TT4, respectively. These findings provided evidence for associations between prenatal PFAS exposure and epigenetic changes in placental TH-related genes.
Collapse
Affiliation(s)
- Zhenzhen Xie
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Longmei Jin
- Minhang Maternal and Child Health Hospital, Shanghai 201100, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Honglei Ji
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Maohua Miao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Xiuxia Song
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Haijun Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Huijia Su
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Wei Yuan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China.
| | - Hong Liang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China.
| |
Collapse
|
2
|
Halloran KM, Saadat N, Pallas B, Vyas AK, Padmanabhan V. Exploratory analysis of differences at the transcriptional interface between the maternal and fetal compartments of the sheep placenta and potential influence of fetal sex. Mol Cell Endocrinol 2025; 603:112546. [PMID: 40222550 DOI: 10.1016/j.mce.2025.112546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025]
Abstract
An understanding of the inner workings of the placenta is imperative to elucidate how the maternal and fetal compartments coordinate to mediate fetal development. The two compartments can be separated and studied before term in sheep, a feat not possible in humans, thus providing a valuable translational model. This study investigated differential expression of gene signaling networks in the maternal and fetal compartments of the placenta and explored the potential influence of fetal sex. On approximately gestational day 120 (term: 147 days), ewes were euthanized and fetuses removed and sexed. Placentomes [n = 5 male, n = 3 female] were collected, and caruncles (maternal) and cotyledons (fetal) were separated and sequenced to assess RNA expression. Analysis revealed 2627 differentially expressed genes (FDR<0.01, abslog2FC ≥ 2) contributing to key transcriptional differences between maternal and fetal compartments, which suggested that the maternal compartment drives extracellular signaling at the interface whereas the fetal compartment controls internal mechanisms crucial for fetal-placental development. X-chromosome inactivation equalized expression of a vast majority of X-linked genes in the fetal compartment. Additionally, the female placenta had more fine-tuned regulation of key pathways for fetal-placental development, such as DNA replication, mRNA surveillance, and RNA transport, compared to males, which had enrichment of metabolic pathways including TCA cycle and galactose metabolism. These findings, in addition to supporting differences in expression in the maternal and fetal placental compartments and the possible influence of fetal sex, offer a transcriptional platform to compare placental perturbations that occur at the maternal-fetal interface that contribute to adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | - Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Arpita K Vyas
- Department of Pediatrics, Washington University St. Louis, MO, USA
| | | |
Collapse
|
3
|
Ebbing C, Halmoy A, Rasmussen S, Mauland KK, Kessler J, Moster D. Umbilical cord length and neurodevelopmental disorders, a national cohort study. PLoS One 2025; 20:e0322444. [PMID: 40267150 PMCID: PMC12017576 DOI: 10.1371/journal.pone.0322444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
INTRODUCTION Adversities in fetal life are known risk factors for neurodevelopmental disorders (NDD). Despite the pivotal role of the umbilical cord, little is known about its associations to later NDD. OBJECTIVE To estimate the associations between umbilical cord length and NDD (Attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), intellectual disability (ID), cerebral palsy (CP), epilepsy, impaired vision or hearing), and whether associations differed by sex. MATERIALS AND METHODS A prospective population-based cohort study including all liveborn singletons in Norway from 1999, through 2013 and followed up through 2019. Data were retrieved from The Medical Birth Registry of Norway and linked with other national health and administrative registries. Exposures were extreme umbilical cord length (empirical percentile <5th or ≥ 95th percentiles). Main outcome measures were NDD (ADHD, ASD, ID, CP, epilepsy, impaired vision or hearing). Associations with umbilical cord length were assessed using logistic regression. RESULTS The cohort consisted of 858,397 births (51.3% boys). We identified 33,370 persons with ADHD (69.8% boys), 10,818 had ASD (76.0% boys), 5538 ID (61.4% boys), 2152 with CP (59.9% boys), 8233 epilepsy (55.0% boys), 900 impaired vision (boys 55.0%), and 11,441 impaired hearing (boys 52.8%). Cord length was positively associated with ADHD (OR 1.15; 95%CI 1.09-1.22), i.e., the risk increased with long cord and decreased with short cord, regardless of sex. A short cord was positively associated with ID (OR 2.42; 95%CI 2.17-2.69), impaired hearing (OR 1.41; 95%CI 1.29-1.54), and epilepsy (OR 1.31; 95%CI 1.18-1.46). CP was associated with both short and long cord (OR 1.31; 95% CI 1.07-1.61 and 1.34, 95%CI 1.13-1.60, respectively). There was no association between cord length and impaired vision. CONCLUSIONS This first population study finds that umbilical cord length is associated with NDD. The findings support the hypothesis that neurodevelopment and development of the umbilical cord share pathways.
Collapse
Affiliation(s)
- Cathrine Ebbing
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Anne Halmoy
- Department of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Svein Rasmussen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Karen K. Mauland
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jørg Kessler
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Dag Moster
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
4
|
Teo SM, Segurado R, Douglass A, McAuliffe FM, Murrin C, Kelleher CC, Phillips CM. Associations between placental parameters and childhood BMI trajectories from birth to nine years of age in the Lifeways Cross-Generation Cohort. Am J Clin Nutr 2025:S0002-9165(25)00144-3. [PMID: 40139274 DOI: 10.1016/j.ajcnut.2025.03.016] [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: 10/03/2024] [Revised: 02/21/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Understanding factors influencing childhood body mass index (BMI) trajectories is crucial for addressing early obesity origins. Placental measures, including placental weight (PW) and birthweight:placental weight (BW:PW) ratio, impact birth outcomes, but their long-term effects on postnatal growth trajectories remain unclear. OBJECTIVES The objectives of this study were to examine associations between placental outcomes and offspring BMI growth over the first 9 y of life and to consider sex-specific and other potential factors affecting growth. METHODS Secondary analysis of longitudinal data from 275 mother-child pairs in the Lifeways Cross-Generational Cohort was examined using linear mixed models. Mothers provided data on age, height, prepregnancy weight, lifestyle, and education. Prepregnancy BMI was calculated. Birth outcomes (length, head circumference, gestational age, PW, and BW) and sex were obtained from hospital records, and the BW:PW ratio was generated. Children's weights and heights measured at birth, 5-, and 9-y follow-up were used to calculate BMI. Maternal age, education, and smoking were adjusted for as confounders in the final model, and interaction terms were used to test moderators. RESULTS In the final model, PW was positively associated with offspring BMI (B: 0.005; 95% CI: 0.004, 0.007). However, this effect diminished over time (B: -0.003; 95% CI: -0.006, -0.001 and B: -0.004; 95% CI: -0.007, -0.001 at age 5 and 9 y, respectively). No significant sex differences or associations with BW:PW ratio were observed with BMI trajectories. Maternal BMI was positively associated with offspring BMI gains, particularly at age 9 y (B: 0.25; 95% CI: 0.13, 0.38). CONCLUSIONS Higher PW is initially associated with higher offspring BMI in a nonsex-specific manner, but its influence diminishes by the age of 9 y. The influence of maternal BMI on offspring BMI at age 9 may reflect the cumulative impact of maternal BMI on offspring BMI trajectories as children approach preadolescence. Future research should include larger, more diverse populations to better understand these associations.
Collapse
Affiliation(s)
- Shevaun M Teo
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Ricardo Segurado
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Alexander Douglass
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Celine Murrin
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Cecily C Kelleher
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Catherine M Phillips
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
5
|
Chevalley T, Dübi M, Fumeaux L, Merli MS, Sarre A, Schaer N, Simeoni U, Yzydorczyk C. Sexual Dimorphism in Cardiometabolic Diseases: From Development to Senescence and Therapeutic Approaches. Cells 2025; 14:467. [PMID: 40136716 PMCID: PMC11941476 DOI: 10.3390/cells14060467] [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: 02/12/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
The global incidence and prevalence of cardiometabolic disorders have risen significantly in recent years. Although lifestyle choices in adulthood play a crucial role in the development of these conditions, it is well established that events occurring early in life can have an important effect. Recent research on cardiometabolic diseases has highlighted the influence of sexual dimorphism on risk factors, underlying mechanisms, and response to therapies. In this narrative review, we summarize the current understanding of sexual dimorphism in cardiovascular and metabolic diseases in the general population and within the framework of the Developmental Origins of Health and Disease (DOHaD) concept. We explore key risk factors and mechanisms, including the influence of genetic and epigenetic factors, placental and embryonic development, maternal nutrition, sex hormones, energy metabolism, microbiota, oxidative stress, cell death, inflammation, endothelial dysfunction, circadian rhythm, and lifestyle factors. Finally, we discuss some of the main therapeutic approaches, responses to which may be influenced by sexual dimorphism, such as antihypertensive and cardiovascular treatments, oxidative stress management, nutrition, cell therapies, and hormone replacement therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine Yzydorczyk
- Developmental Origins of Health and Disease (DOHaD) Laboratory, Division of Pediatrics, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (T.C.); (M.D.); (L.F.); (M.S.M.); (A.S.); (N.S.)
| |
Collapse
|
6
|
Beck IH, Grøntved A, Palm CVB, Batzella E, Sigvaldsen A, Dalgård C, Jensen RC, Nielsen C, Halldorsson TI, Jensen TK. Prenatal PFAS exposure associates with DXA assessed markers of adiposity in 7-year-old children from the Odense Child Cohort. ENVIRONMENTAL RESEARCH 2025; 275:121394. [PMID: 40086573 DOI: 10.1016/j.envres.2025.121394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/16/2025]
Abstract
The global increase in childhood overweight and obesity presents significant public health concerns due to its long-term health implications. Emerging evidence suggests that exposure to endocrine disrupting chemicals, such as per- and polyfluoroalkylated substances (PFAS), may be obesogenic and contribute to adiposity. This study aimed to investigate the association between prenatal PFAS exposure and markers of adiposity in 7-year-old children, focusing on potential sex-specific differences. Data was analyzed from 881 mother-child pairs in the Odense Child Cohort, Denmark. Maternal serum concentrations of perfluorohexane sulfonic acid (PFHxS), perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), and perfluorodecanoic acid (PFDA) were measured in early pregnancy. At age 7, body composition, including body mass index (BMI), lean mass and fat distribution (total, gynoid, and android), was assessed using dual-energy X-ray absorptiometry (DXA). The median (25th;75th percentile) concentrations of PFHxS, PFOS, PFOA, PFNA, and PFDA were 0.4 (0.2;0.5), 7.6 (5.6;10.4), 1.7 (1.1;2.3), 0.6 (0.5;0.8), and 0.3 (0.2;0.4) ng/mL, respectively. Multiple linear regressions were used to assess sex specific associations between maternal PFAS concentrations and markers of adiposity. In girls, 1 ng/mL increase in maternal PFOA was associated with 2.0 % (95 % confidence interval: 0.3; 3.7) increase in total fat, 1.3 % (-0.3; 2.9) increase in gynoid fat, and 3.8 % (0.6; 7.0) increase in android fat. Associations for PFNA and PFDA followed similar trends, whereas higher maternal PFOS concentrations were associated with lower BMI among both girls and boys. These findings suggest that prenatal exposure to certain PFAS may influence the accumulation of excess fat in girls. Our findings highlight the importance of studying sex specific differences and using accurate measures of body composition as BMI may not adequately reflect body fat in children during growth.
Collapse
Affiliation(s)
- Iben Have Beck
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Kløvervænget 23C, Odense C, 5000, Denmark.
| | - Anders Grøntved
- Department of Sports Science and Clinical Biomechanics, Research Unit for Exercise Epidemiology, Centre of Research in Childhood Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Camilla V B Palm
- Department of Endocrinology and Metabolism, Odense University Hospital, Kløvervænget 6, Odense C, 5000, Denmark
| | - Erich Batzella
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Annika Sigvaldsen
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Kløvervænget 23C, Odense C, 5000, Denmark
| | - Christine Dalgård
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Richard Christian Jensen
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark; Department of Endocrinology and Metabolism, Odense University Hospital, Kløvervænget 6, Odense C, 5000, Denmark
| | - Christel Nielsen
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark; Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Scheelevägen 8, 223 81, Lund, Sweden
| | - Thorhallur I Halldorsson
- Center for Fetal Programming, Department of Epidemiology Research, Statens Serum Institut, Artillerivej 5, DK-2300, København S., Denmark; Faculty of Food Science and Nutrition, School of Health Sciences, University of Iceland, Sæmundargata 12, 102, Reykjavík, Iceland
| | - Tina Kold Jensen
- Department Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Kløvervænget 23C, Odense C, 5000, Denmark; Open Patient Data Explorative Network, Odense University Hospital, J. B. Winsløws Vej 21, 3. sal, DK-5000, Odense, Denmark
| |
Collapse
|
7
|
Maurer SV, Hing BWQ, Lussier S, Radhakrishna S, Davis JLB, Abbott PW, Michaelson JJ, Stevens HE. Prenatal stress alters mouse offspring dorsal striatal development and placental function in sex-specific ways. J Psychiatr Res 2025; 182:149-160. [PMID: 39809011 PMCID: PMC11959308 DOI: 10.1016/j.jpsychires.2024.12.048] [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: 11/15/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Prenatal stress is a risk factor for neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD). However, how early stress modification of brain development contributes to this pathophysiology is poorly understood. Ventral forebrain regions such as dorsal striatum are of particular interest: dorsal striatum modulates movement and cognition, is altered in NDDs, and has a primarily GABAergic population. Here, we examine effects of prenatal stress on adult movement, cognition, and dorsal striatum neurobiology in mice using striatal-dependent behavioral assays, immunohistochemistry, embryonic ventral forebrain transcriptomics, and placental transcriptomics. We found prenatal stress affected adult procedural, habit, and reversal learning in sex-specific ways. Stress also increased adult dorsal striatal GABAergic neurons - an effect largely driven by males. We sought to examine the developmental origins of these adult brain changes. We found similar sex-specific dorsal striatal cellular changes in earlier points of development. The dorsal striatum primordium--embryonic ventral forebrain-showed that prenatal stress increased DNA replication and cell cycle pathways in male but not female transcriptomics and cellular biology. Unique signatures may have arisen from male-female placental differences. Stress-induced placental transcriptomics showed upregulated morphogenesis pathways in males while females downregulated morphogenic, hormonal, and cellular response pathways. Our findings suggest that prenatal stress could affect placenta function and also alter the GABAergic population of dorsal striatum differentially between the sexes.
Collapse
Affiliation(s)
- Sara V Maurer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Benjamin W Q Hing
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Stephanie Lussier
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SL is now with Moderna, USA
| | - Sreya Radhakrishna
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SR is now at Albert Einstein College of Medicine, USA
| | - Jada L B Davis
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Parker W Abbott
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
8
|
Rak K, Godyla-Jabłoński M, Bronkowska M. Sex-specific association of immunological markers in CS-delivered newborns with pre-pregnancy body mass index and gestational weight gain of mothers. Sci Rep 2025; 15:3074. [PMID: 39856119 PMCID: PMC11760347 DOI: 10.1038/s41598-025-85711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Just as overweight and obesity may impair immunity, excessive body weight-related parameters of women in the pre-conception period and during pregnancy are possible detrimental factors for fetal programming of the immune system in their offspring. We investigated the relationship of pre-pregnancy body mass index (pBMI) and gestational weight gain (GWG) of mothers with the placental transport rate (PTR) of IgG antibodies and antineutrophil cytoplasmatic antibodies against lactoferrin (Lf-ANCA) and their concentration in umbilical cord blood serum (UCS), verifying the sex-specificity of this relationship. The examined group of this cross-sectional pilot study consisted of 101 pregnant women and their healthy CS-delivered newborn children. The concentration of antibodies in maternal serum (MS) and UCS were determined by ELISA method. PTR was assessed as a ratio of the concentration of antibodies in the UCS and MS. A significantly lower PTR of IgG and their concentration in the UCS were demonstrated in newborns of mothers with an excessive pBMI compared to those with pBMI < 25 and the association was more pronounced in male newborns. The lowest PTR of beneficial IgG and their concentration in the UCS as well as the highest PTR of detrimental Lf-ANCA and their concentration in UCS were observed in newborns born to mothers with co-occurrence of both an excessive pBMI and GWG. It seems that maternal preconception overweight and obesity along with an excessive GWG can be a predictor of unfavorable immune effects in fetuses. Further studies are needed to explain the role of maternal weight-related parameters in the development of immunological health of their offspring.
Collapse
Affiliation(s)
- Karolina Rak
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, Wrocław, 51-630, Poland.
| | - Michaela Godyla-Jabłoński
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, Wrocław, 51-630, Poland
| | - Monika Bronkowska
- Institute of Health Sciences, Collegium Salutis Humanae, University of Opole, Opole, 45-060, Poland
| |
Collapse
|
9
|
Li ZH, Hu CY, Dai SW, Ma HY, Zhang SY, Sun C, Li JH, Huang K, Chen ML, Gao GP, Zhang XJ. Sex-specific associations between maternal exposure to metal mixtures and fetal growth trajectories: A prospective birth cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178291. [PMID: 39733573 DOI: 10.1016/j.scitotenv.2024.178291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND The associations of prenatal metals exposure with birth outcomes have been widely assessed. However, evidence on the associations between metal mixtures and fetal intrauterine growth trajectories is scarce. OBJECTIVES This study aimed to explore the associations of metal mixtures with fetal intrauterine growth trajectories overall and by sex. METHODS We analyzed data from the Ma'anshan birth cohort, which included a total of 1041 pregnant woman. The concentrations of 12 metals in maternal blood were measured during early pregnancy, and fetal intrauterine growth indicators were standardized and assessed at 16, 23, 30, 34, and 38 weeks of gestation. We used generalized linear regression and linear mixed models to identify the key fetal growth indicator (biparietal diameter (BPD)), and applied GBTM to characterize BPD SD-scores trajectories. To further assess the individual and combined effects of metals, we conducted multivariable logistic regression and repeated holdouts weighted quantile sum (WQS) regression analyses, respectively. Finally, we performed a sex-stratified analysis to explore sex-specific associations. RESULTS The sex-stratified multivariable logistic regression analysis indicated that in male fetal, cobalt (Co) (OR: 0.60, 95 % CI: 0.38, 0.92) was negatively associated with the high-growth BPD-SD scores trajectory. In contrast, Co (OR: 2.39, 95 % CI: 1.40, 4.45) showed a positive association in female fetal. Results from the WQS showed that early pregnancy metal mixture exposure was associated with BPD-SD scores at 16, 34, and 38 weeks in female fetal. The results highlighted Zn and Co as key metals associated with high-growth BPD SD-scores trajectory. We also identified a significant interaction between early pregnancy metal mixtures and sex on high-growth BPD SD-scores trajectories. The WQS*sex interaction term had a mean odds ratio of 1.271 (95 % CI: 1.027, 1.619). CONCLUSION This study suggests that exposure to prenatal metal mixtures affects fetal intrauterine growth trajectories with sexual dimorphism.
Collapse
Affiliation(s)
- Zhen-Hua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Cheng-Yang Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Department of Humanistic Medicine, School of Humanistic Medicine, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Si-Wei Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Hui-Ya Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Management & Checkup Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, Anhui, China
| | - Si-Yu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Chen Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Jia-Hui Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Kai Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Department of Hospital Infection Prevention and Control, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, China
| | - Mao-Lin Chen
- Department of Gynecology and Obstetrics, Ma'anshan Maternal and Child Health Hospital, Ma'anshan 243000, China
| | - Guo-Peng Gao
- Department of Child Health Care, Ma'anshan Maternal and Child Health Hospital, Ma'anshan 243000, China
| | - Xiu-Jun Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
10
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. A transcriptomic comparison of in vitro models of the human placenta. Placenta 2025; 159:52-61. [PMID: 39637677 PMCID: PMC11857522 DOI: 10.1016/j.placenta.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. The aim of this research was to compare the transcriptomes of common in vitro models of the human placenta compared to bulk human placental tissue. METHODS We performed differential gene expression analysis on publicly available transcriptomic data from 7 in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, Villous Explants, and Trophoblast Stem Cells) and compared to bulk placental tissue from 2 cohort studies (CANDLE and GAPPS) or individual trophoblast cell types derived from bulk placental tissue. RESULTS All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs = 10,624), and the individual trophoblast cell types (Average DEGs = 5413), indicating that there are vast differences in gene expression. Hierarchical clustering identified 54 gene clusters with distinct expression profiles across placental models, with 23 clusters enriched for specific KEGG pathways. Placental cell lines were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as female origin, while JEG-3, JAR, and BeWo cells are of male origin. DISCUSSION None of the models were a close approximation of the human bulk placental transcriptome, highlighting the challenges with model selection. To enable appropriate model selection, we adapted our data into a web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)".
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Evan J Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, 98101, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| |
Collapse
|
11
|
Di Pietrantonio N, Sánchez-Ceinos J, Shumliakivska M, Rakow A, Mandatori D, Di Tomo P, Formoso G, Bonfini T, Baldassarre MPA, Sennström M, Almahmeed W, Pandolfi A, Cosentino F. The inflammatory and oxidative phenotype of gestational diabetes is epigenetically transmitted to the offspring: role of methyltransferase MLL1-induced H3K4me3. Eur Heart J 2024; 45:5171-5185. [PMID: 39471481 DOI: 10.1093/eurheartj/ehae688] [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: 02/08/2024] [Revised: 06/16/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND AND AIMS Hyperglycaemia during gestational diabetes (GD) predisposes women and their offspring to later cardiometabolic disease. The hyperglycaemia-mediated epigenetic changes remain to be elucidated. Methyltransferase MLL1-induced trimethylation of histone 3 at lysine 4 (H3K4me3) activates inflammatory and oxidative phenotype. This epigenetic mark in GD women and its transmission to the offspring were investigated. METHODS Peripheral blood mononuclear cells (PBMC) were collected from GD and control (C) women and also from adolescents born to women of both groups. Endothelial human umbilical vein endothelial cells (HUVEC) and cord blood mononuclear cells (CBMC) were from umbilical cords. The NF-κBp65 and NOX4 expressions were investigated by reverse transcription quantitative polymerase chain reaction and immunofluorescence (IF). MLL1 and H3K4me3 were investigated by immunoblotting and IF. H3K4me3 on NF-κBp65 and NOX4 promoters was studied by chromatin immunoprecipitation. Superoxide anion generation was measured by electron spin resonance spectroscopy. Plasma cytokines were measured by enzyme-linked immunosorbent assay. To investigate the role of MLL1, HUVEC were exposed to inhibitor MM102 or siRNA transfection. RESULTS PBMC, CBMC, and HUVEC showed an increase of NF-κBp65, IL-6, ICAM-1, MCP-1, and VCAM-1 mRNAs. These findings were associated with H3K4me3 enrichment in the promoter of NF-κBp65. Elevated H3K4me3 and cytokine levels were observed in GD adolescents. MLL1 drives H3K4me3 not only on NF-kB p65, but also on NOX4 promoter. Inhibition of MLL1 blunted NF-κBp65 and NOX4 by modulating inflammatory and oxidative phenotype. CONCLUSIONS Such proof-of-concept study shows persistence of MLL1-dependent H3K4me3 in offspring born to GD women, suggesting an epigenetic-driven transmission of maternal phenotype. These findings may pave the way for pharmacological reprogramming of adverse histone modifications to mitigate abnormal phenotypes underlying early ASCVD.
Collapse
Affiliation(s)
- Nadia Di Pietrantonio
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Julia Sánchez-Ceinos
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Mariana Shumliakivska
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Alexander Rakow
- Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Pamela Di Tomo
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Gloria Formoso
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Tiziana Bonfini
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Maria Pompea Antonia Baldassarre
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Maria Sennström
- Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm 171 76, Sweden
| |
Collapse
|
12
|
Aljabali SM, Pai S, Teperino R. Paternal impact on the developmental programming of sexual dimorphism. Front Cell Dev Biol 2024; 12:1520783. [PMID: 39712575 PMCID: PMC11659275 DOI: 10.3389/fcell.2024.1520783] [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: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Sexual dimorphism involves distinct anatomical, physiological, behavioral, and developmental differences between males and females of the same species, influenced by factors prior to conception and during early development. These sex-specific traits contribute to varied phenotypes and individual disease risks within and across generations and understanding them is essential in mammalian studies. Hormones, sex chromosomes, and imprinted genes drive this dimorphism, with over half of quantitative traits in wildtype mice showing sex-based variation. This review focuses on the impact of paternal non-genetic factors on sexual dimorphism. We synthesize current research on how paternal health before conception affects offspring phenotypes in a sex-specific manner, examining mechanisms such as DNA methylation, paternally imprinted genes, sperm RNA, and seminal plasma. Additionally, we explore how paternal influences indirectly shape offspring through maternal behavior, uterine environment, and placental changes, affecting males and females differently. We propose mechanisms modulating sexual dimorphism during development, underscoring the need for sex-specific documentation in animal studies.
Collapse
Affiliation(s)
- Shefa’ M. Aljabali
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| | - Shruta Pai
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Munich GmbH, German Research Center for Environmental Health, Neuherberg, Germany
- DZD – German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
13
|
Paparini DE, Grasso E, Aguilera F, Arslanian MA, Lella V, Lara B, Schafir A, Gori S, Merech F, Hauk V, Schuster C, Martí M, Meller C, Ramhorst R, Vota D, Leirós CP. Sex-specific phenotypical, functional and metabolic profiles of human term placenta macrophages. Biol Sex Differ 2024; 15:80. [PMID: 39420346 PMCID: PMC11484421 DOI: 10.1186/s13293-024-00652-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Placental macrophages, Hofbauer cells (HBC) are the only fetal immune cell population within the stroma of healthy placenta along pregnancy. They are central players in maintaining immune tolerance during pregnancy. Immunometabolism emerged a few years ago as a new field that integrates cellular metabolism with immune responses, however, the immunometabolism of HBC has not been explored yet. Here we studied the sex-specific differences in the phenotypic, functional and immunometabolic profile of HBC. METHODS HBC were isolated from human term placentas (N = 31, 16 from male and 15 female neonates). Ex vivo assays were carried out to assess active metabolic and endoplasmic reticulum stress pathways by flow cytometry, confocal microscopy, gene expression and in silico approaches. RESULTS HBC from female placentas displayed a stronger M2 phenotype accompanied by high rates of efferocytosis majorly sustained on lipid metabolism. On the other hand, male HBC expressed a weaker M2 phenotype with higher glycolytic metabolism. LPS stimulation reinforced the glycolytic metabolism in male but not in female HBC. Physiological endoplasmic reticulum stress activates IRE-1 differently, since its pharmacological inhibition increased lipid mobilization, accumulation and efferocytosis only in female HBC. Moreover, differential sex-associated pathways accompanying the phenotypic and functional profiles of HBC appeared related to the placental villi environment. CONCLUSIONS These results support sex-associated effects on the immunometabolism of the HBC and adds another layer of complexity to the intricate maternal-fetal immune interaction.
Collapse
Affiliation(s)
- Daniel E Paparini
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Esteban Grasso
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Franco Aguilera
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | - Victoria Lella
- Obstetric Service, Hospital Italiano, Buenos Aires, Argentina
| | - Brenda Lara
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana Schafir
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Soledad Gori
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Fátima Merech
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudio Schuster
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marcelo Martí
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cesar Meller
- Obstetric Service, Hospital Italiano, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
14
|
Sánchez-Rubio M, Abarzúa-Catalán L, Del Valle A, Méndez-Ruette M, Salazar N, Sigala J, Sandoval S, Godoy MI, Luarte A, Monteiro LJ, Romero R, Choolani MA, Wyneken Ú, Illanes SE, Bátiz LF. Maternal stress during pregnancy alters circulating small extracellular vesicles and enhances their targeting to the placenta and fetus. Biol Res 2024; 57:70. [PMID: 39342314 PMCID: PMC11438166 DOI: 10.1186/s40659-024-00548-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Maternal psychological distress during pregnancy can negatively impact fetal development, resulting in long-lasting consequences for the offspring. These effects show a sex bias. The mechanisms whereby prenatal stress induces functional and/or structural changes in the placental-fetal unit remain poorly understood. Maternal circulating small extracellular vesicles (sEVs) are good candidates to act as "stress signals" in mother-to-fetus communication. Using a repetitive restraint-based rat model of prenatal stress, we examined circulating maternal sEVs under stress conditions and tested whether they could target placental-fetal tissues. RESULTS Our mild chronic maternal stress during pregnancy paradigm induced anhedonic-like behavior in pregnant dams and led to intrauterine growth restriction (IUGR), particularly in male fetuses and placentas. The concentration and cargo of maternal circulating sEVs changed under stress conditions. Specifically, there was a significant reduction in neuron-enriched proteins and a significant increase in astrocyte-enriched proteins in blood-borne sEVs from stressed dams. To study the effect of repetitive restraint stress on the biodistribution of maternal circulating sEVs in the fetoplacental unit, sEVs from pregnant dams exposed to stress or control protocol were labeled with DiR fluorescent die and injected into pregnant females previously exposed to control or stress protocol. Remarkably, maternal circulating sEVs target placental/fetal tissues and, under stress conditions, fetal tissues are more receptive to sEVs. CONCLUSION Our results suggest that maternal circulating sEVs can act as novel mediators/modulators of mother-to-fetus stress communication. Further studies are needed to identify placental/fetal cellular targets of maternal sEVs and characterize their contribution to stress-induced sex-specific placental and fetal changes.
Collapse
Affiliation(s)
- Mario Sánchez-Rubio
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Lorena Abarzúa-Catalán
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Ana Del Valle
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Maxs Méndez-Ruette
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- PhD Program in Biomedicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Natalia Salazar
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Jacinta Sigala
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Soledad Sandoval
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - María Inés Godoy
- Department of Educational Assessment, Measurement, and Registry, Universidad de Chile, Santiago, Chile
| | - Alejandro Luarte
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Lara J Monteiro
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Research Program in Biology of Reproduction, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Detroit, and Maryland, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Mahesh A Choolani
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Úrsula Wyneken
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Sebastián E Illanes
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Research Program in Biology of Reproduction, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile.
- Department of Obstetrics and Gynecology, School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Facultad de Medicina, Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| | - Luis Federico Bátiz
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Facultad de Medicina, Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| |
Collapse
|
15
|
Zhou J, Teng Y, Ouyang J, Wu P, Tong J, Gao G, Yan S, Tao F, Huang K. Associations of Placental Inflammation and Oxidative Stress Biomarkers with Glucolipid Metabolism in Children: A Birth Cohort Study in China. J Am Heart Assoc 2024; 13:e035754. [PMID: 39206740 PMCID: PMC11646502 DOI: 10.1161/jaha.124.035754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The maternal intrauterine immune environment may affect offspring long-term health. We aimed to investigate the association between the intrauterine placental immunological milieu and glycolipid metabolic health in children. METHODS AND RESULTS This study enrolled 1803 mother-child pairs from the Ma'anshan birth cohort (2013-2014). Placental mRNA expression of inflammatory cytokines (interleukin-1β [IL-1β], IL-10, monocyte chemoattractant protein-1, tumor necrosis factor-α, IL-4, IL-6, IL-8, C-reactive protein, and interferon-γ) and oxidative stress biomarkers (heme oxygenase-1, hypoxia-inducible factor-1alpha, and glucose-related protein 78) was quantified using real-time quantitative polymerase chain reaction. Fasting blood glucose, insulin, triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and total cholesterol were assessed at 5 to 6 years old. Statistical analyses included multiple linear regression, binary logistic regression, restricted cubic spline model, and the Bayesian kernel machine regression model. Placental inflammatory cytokines (IL-1β, monocyte chemoattractant protein-1, C-reactive protein, IL-6, IL-8, IL-10) and oxidative stress biomarkers (heme oxygenase-1, hypoxia-inducible factor-1alpha, glucose-related protein 78) showed positive associations with children's fasting blood glucose levels. Heme oxygenase-1 and glucose-related protein 78 exhibited negative correlations with children's fasting insulin levels. Elevated IL-6, heme oxygenase-1, hypoxia-inducible factor-1alpha, and glucose-related protein 78 were associated with increased risk of prediabetes in children. Overall upregulation of placental proinflammatory cytokines and oxidative stress factors mRNA expression correlated with higher prediabetes risk in children. Bayesian kernel machine regression analysis indicated a joint positive effect of the 12 placental inflammation and oxidative stress mixtures on children's risk of high fasting blood glucose. CONCLUSIONS This exploratory study underscores significant correlations between maternal intrauterine placental inflammation, oxidative stress markers, and offspring fasting blood glucose and insulin levels. These findings highlight the potential role of intrauterine holistic immunity in shaping offspring glucose metabolism health.
Collapse
Affiliation(s)
- Jixing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Yuzhu Teng
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Jiajun Ouyang
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Penggui Wu
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Guopeng Gao
- Maternal and Child Health Care Center of Ma’anshanAnhuiChina
| | - Shuangqin Yan
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Maternal and Child Health Care Center of Ma’anshanAnhuiChina
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| |
Collapse
|
16
|
Shah RG, Salafia CM, Girardi T, Rukat C, Brunner J, Barrett ES, O'Connor TG, Misra DP, Miller RK. Maternal affective symptoms and sleep quality have sex-specific associations with placental topography. J Affect Disord 2024; 360:62-70. [PMID: 38806063 DOI: 10.1016/j.jad.2024.05.108] [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: 11/22/2023] [Revised: 04/10/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND The impacts of prenatal maternal affective symptoms on the placental structure are not well-established. Employing Geographic Information System (GIS) spatial autocorrelation, Moran's I, can help characterize placental thickness uniformity/variability and evaluate the impacts of maternal distress on placental topography. METHODS This study (N = 126) utilized cohort data on prenatal maternal affective symptoms and placental 2D and 3D morphology. Prenatal maternal depression, stress, anxiety and sleep quality were scored for each trimester using the Edinburgh Postnatal Depression Scale (EPDS), Stressful Life Event Scale (SLE), Penn State Worry Questionnaire (PSWQ), and Pittsburgh Sleep Quality Index (PSQI), respectively. Placental shape was divided into Voronoi cells and thickness variability among these cells was computed using Moran's I for 4-nearest neighbors and neighbors within a 10 cm radius. Sex-stratified Spearman correlations and linear regression were used to study associations between mean placental thickness, placental GIS variables, placental weight and the average score of each maternal variable. RESULTS For mothers carrying boys, poor sleep was associated with higher mean thickness (r = 0.308,p = 0.035) and lower placental thickness uniformity (r = -0.36,p = 0.012). Lower placental weight (r = 0.395,p = 0.003), higher maternal depression (r = -0.318,p = 0.019) and worry/anxiety (r = -0.362,p = 0.007) were associated with lower placental thickness uniformity for mothers carrying girls. LIMITATIONS The study is exploratory and not all GIS models were developed. Excluding high-risk pregnancies prevented investigating pregnancy complications related hypotheses. A larger sample size is needed for greater confidence for clinical application. CONCLUSIONS Placental topography can be studied using GIS theory and has shown that prenatal maternal affective symptoms and sleep have sex-specific associations with placental thickness.
Collapse
Affiliation(s)
- Ruchit G Shah
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA.
| | - Carolyn M Salafia
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Theresa Girardi
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Cate Rukat
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Jessica Brunner
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health; Environmental and Occupational Health Sciences Institute, Piscataway, USA
| | - Thomas G O'Connor
- Departments of Psychiatry, Obstetrics/Gynecology, Pediatrics, University of Rochester, School of Medicine and Dentistry, Rochester, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, Michigan State University, MI, USA
| | - Richard K Miller
- Departments of Obstetrics and Gynecology, Environmental Medicine, Pathology, and Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, USA
| |
Collapse
|
17
|
Xie Y, Zhao F, Wang Y, Borowski S, Freitag N, Tirado-Gonzalez I, Hofsink N, Matschl U, Plösch T, Garcia MG, Blois SM. Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis. Cell Death Dis 2024; 15:575. [PMID: 39117607 PMCID: PMC11310209 DOI: 10.1038/s41419-024-06962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis.
Collapse
Affiliation(s)
- Yiran Xie
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fangqi Zhao
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yiru Wang
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sophia Borowski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and Institute of Biochemistry, Berlin, Germany and Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Nancy Freitag
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and Institute of Biochemistry, Berlin, Germany and Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Irene Tirado-Gonzalez
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Urte Matschl
- Department Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences, Carlvon Ossietzky University Oldenburg, Oldenburg, Germany
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Hodge KM, Zhabotynsky V, Burt AA, Carter BS, Fry RC, Helderman J, Hofheimer JA, McGowan EC, Neal CR, Pastyrnak SL, Smith LM, DellaGrotta SA, Dansereau LM, Lester BM, Marsit CJ, O'Shea TM, Everson TM. Epigenetic associations in HPA axis genes related to bronchopulmonary dysplasia and antenatal steroids. Pediatr Res 2024; 96:510-518. [PMID: 38480856 DOI: 10.1038/s41390-024-03116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/26/2024] [Accepted: 02/17/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD), a common morbidity among very preterm infants, is associated with chronic disease and neurodevelopmental impairments. A hypothesized mechanism for these outcomes lies in altered glucocorticoid (GC) activity. We hypothesized that BPD and its treatments may result in epigenetic differences in the hypothalamic-pituitary-adrenal (HPA) axis, which is modulated by GC, and could be ascertained using an established GC risk score and DNA methylation (DNAm) of HPA axis genes. METHODS DNAm was quantified from buccal tissue (ECHO-NOVI) and from neonatal blood spots (ELGAN ECHO) via the EPIC microarray. Prenatal maternal characteristics, pregnancy complication, and neonatal medical complication data were collected from medical record review and maternal interviews. RESULTS The GC score was not associated with steroid exposure or BPD. However, six HPA genes involved in stress response regulation demonstrated differential methylation with antenatal steroid exposure; two CpGs within FKBP5 and POMC were differentially methylated with BPD severity. These findings were sex-specific in both cohorts; males had greater magnitude of differential methylation within these genes. CONCLUSIONS These findings suggest that BPD severity and antenatal steroids are associated with DNAm at some HPA genes in very preterm infants and the effects appear to be sex-, tissue-, and age-specific. IMPACT This study addresses bronchopulmonary dysplasia (BPD), an important health outcome among preterm neonates, and interrogates a commonly studied pathway, the hypothalamic-pituitary-adrenal (HPA) axis. The combination of BPD, the HPA axis, and epigenetic markers has not been previously reported. In this study, we found that BPD itself was not associated with epigenetic responses in the HPA axis in infants born very preterm; however, antenatal treatment with steroids was associated with epigenetic responses.
Collapse
Affiliation(s)
- Kenyaita M Hodge
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Vasyl Zhabotynsky
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amber A Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Brian S Carter
- Department of Pediatrics-Neonatology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jennifer Helderman
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Julie A Hofheimer
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Elisabeth C McGowan
- Department of Pediatrics, Warren Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI, USA
| | - Charles R Neal
- Department of Pediatrics, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | - Steven L Pastyrnak
- Department of Pediatrics, Spectrum Health-Helen Devos Hospital, Grand Rapids, MI, USA
| | - Lynne M Smith
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sheri A DellaGrotta
- Brown Center for the Study of Children at Risk, Women and Infants Hospital, Providence, RI, USA
| | - Lynne M Dansereau
- Brown Center for the Study of Children at Risk, Women and Infants Hospital, Providence, RI, USA
| | - Barry M Lester
- Department of Pediatrics, Warren Alpert Medical School of Brown University and Women and Infants Hospital, Providence, RI, USA
- Brown Center for the Study of Children at Risk, Women and Infants Hospital, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - T Michael O'Shea
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Todd M Everson
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Siwakoti RC, Park S, Ferguson KK, Hao W, Cantonwine DE, Mukherjee B, McElrath TF, Meeker JD. Prenatal per- and polyfluoroalkyl substances (PFAS) and maternal oxidative stress: Evidence from the LIFECODES study. CHEMOSPHERE 2024; 360:142363. [PMID: 38768789 PMCID: PMC11487489 DOI: 10.1016/j.chemosphere.2024.142363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals linked to adverse pregnancy outcomes. Although their underlying biological mechanisms are not fully understood, evidence suggests PFAS may disrupt endocrine functions and contribute to oxidative stress (OS) and inflammation. OBJECTIVE We examined associations between early pregnancy PFAS exposure and OS biomarkers, exploring potential effect modifications by fetal sex and maternal race. METHODS We used data from 469 LIFECODES participants with measured plasma PFAS (median 10 weeks gestation) and repeated measures (median 10, 18, 26, and 35 weeks gestation) of urinary OS biomarkers [8-iso-prostaglandin-F2α (8-isoprostane) and 8-hydroxydeoxyguanosine (8-OHdG)]. Protein damage biomarkers (chlorotyrosine, dityrosine, and nitrotyrosine) were additionally measured in plasma from a subset (N = 167) during the third visit. Associations between each PFAS and OS biomarkers were examined using linear mixed-effects models and multivariable linear regressions, adjusting for potential confounders, including maternal age, race, education level, pre-pregnancy BMI, insurance status, and parity. Effect modifications were evaluated by including an interaction term between each PFAS and fetal sex or maternal race in the models. RESULTS We observed significant positive associations between PFOS and 8-isoprostane, with a 9.68% increase in 8-isoprostane levels (95% CI: 0.10%, 20.18%) per interquartile range increase in PFOS. In contrast, PFUA was negatively associated [9.32% (95% CI: -17.68%, -0.11%)], while there were suggestive positive associations for MPAH and PFOA with 8-isoprostane. The associations of several PFAS with 8-OHdG varied by fetal sex, showing generally positive trends in women who delivered females, but negative or null in those who delivered males. No significant effect modification by maternal race was observed. CONCLUSIONS This study provides evidence linking PFAS exposure to OS during pregnancy, with potential sex-specific effects of certain PFAS on 8-OHdG. Further research should explore additional OS/inflammatory biomarkers and assess the modifying effects of dietary and behavioral patterns across diverse populations.
Collapse
Affiliation(s)
- Ram C Siwakoti
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Seonyoung Park
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kelly K Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Wei Hao
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - David E Cantonwine
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John D Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Paquette A, Ahuna K, Hwang YM, Pearl J, Liao H, Shannon P, Kadam L, Lapehn S, Bucher M, Roper R, Funk C, MacDonald J, Bammler T, Baloni P, Brockway H, Mason WA, Bush N, Lewinn KZ, Karr CJ, Stamatoyannopoulos J, Muglia LJ, Jones H, Sadovsky Y, Myatt L, Sathyanarayana S, Price ND. A genome scale transcriptional regulatory model of the human placenta. SCIENCE ADVANCES 2024; 10:eadf3411. [PMID: 38941464 PMCID: PMC11212735 DOI: 10.1126/sciadv.adf3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/28/2024] [Indexed: 06/30/2024]
Abstract
Gene regulation is essential to placental function and fetal development. We built a genome-scale transcriptional regulatory network (TRN) of the human placenta using digital genomic footprinting and transcriptomic data. We integrated 475 transcriptomes and 12 DNase hypersensitivity datasets from placental samples to globally and quantitatively map transcription factor (TF)-target gene interactions. In an independent dataset, the TRN model predicted target gene expression with an out-of-sample R2 greater than 0.25 for 73% of target genes. We performed siRNA knockdowns of four TFs and achieved concordance between the predicted gene targets in our TRN and differences in expression of knockdowns with an accuracy of >0.7 for three of the four TFs. Our final model contained 113,158 interactions across 391 TFs and 7712 target genes and is publicly available. We identified 29 TFs which were significantly enriched as regulators for genes previously associated with preterm birth, and eight of these TFs were decreased in preterm placentas.
Collapse
Affiliation(s)
- Alison Paquette
- University of Washington, Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Kylia Ahuna
- Oregon Health and Sciences University, Portland, OR, USA
| | | | | | - Hanna Liao
- University of Washington, Seattle, WA, USA
| | | | - Leena Kadam
- Oregon Health and Sciences University, Portland, OR, USA
| | | | - Matthew Bucher
- Oregon Health and Sciences University, Portland, OR, USA
| | - Ryan Roper
- Institute for Systems Biology, Seattle, WA, USA
| | - Cory Funk
- Institute for Systems Biology, Seattle, WA, USA
| | | | | | | | - Heather Brockway
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - W. Alex Mason
- University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Nicole Bush
- University of California San Francisco, San Francisco, CA, USA
| | - Kaja Z. Lewinn
- University of California San Francisco, San Francisco, CA, USA
| | | | | | - Louis J. Muglia
- The Burroughs Wellcome Fund, Research Triangle Park, NC, USA
- Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Yoel Sadovsky
- Magee Womens Research Institute, Pittsburgh, PA, USA
- University of Pittsburgh, Pittsburgh, PA, USA
| | - Leslie Myatt
- Oregon Health and Sciences University, Portland, OR, USA
| | - Sheela Sathyanarayana
- University of Washington, Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Nathan D. Price
- Institute for Systems Biology, Seattle, WA, USA
- Thorne HealthTech, New York City, NY, USA
| |
Collapse
|
21
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. Transcriptomic comparison of in vitro models of the human placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598695. [PMID: 38915703 PMCID: PMC11195179 DOI: 10.1101/2024.06.14.598695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Studying the human placenta through in vitro cell culture methods is necessary due to limited access and amenability of human placental tissue to certain experimental methods as well as distinct anatomical and physiological differences between animal and human placentas. Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. Therefore, the aim of this research was to create a comprehensive transcriptomic comparison of common in vitro models of the human placenta compared to bulk placental tissue from the CANDLE and GAPPS cohorts (N=1083). We performed differential gene expression analysis on publicly available RNA sequencing data from 6 common in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, and Villous Explants) and compared to CANDLE and GAPPS bulk placental tissue or cytotrophoblast, syncytiotrophoblast, and extravillous trophoblast cell types derived from bulk placental tissue. All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs=10,873), and the individual trophoblast cell types (Average DEGs=5,346), indicating that there are vast differences in gene expression compared to bulk and cell-type specific human placental tissue. Hierarchical clustering identified 53 gene clusters with distinct expression profiles across placental models, with 22 clusters enriched for specific KEGG pathways, 7 clusters enriched for high-expression placental genes, and 7 clusters enriched for absorption, distribution, metabolism, and excretion genes. In vitro placental models were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as being of female origin, while JEG-3, JAR, and BeWo cells are of male origin. Overall, none of the models were a close approximation of the transcriptome of bulk human placental tissue, highlighting the challenges with model selection. To enable researchers to select appropriate models, we have compiled data on differential gene expression, clustering, and fetal sex into an accessible web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)" which can be utilized by researchers to make informed decisions about their selection of in vitro placental models.
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Evan J. Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143 United States
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 55905 United States
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202 United States
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
- Center for Child Health, Behavior and Development, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98101 United States
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
| |
Collapse
|
22
|
Freire T, Pulpitel T, Clark X, Mackay F, Raubenheimer D, Simpson SJ, Solon-Biet SM, Crean AJ. The effects of paternal dietary fat versus sugar on offspring body composition and anxiety-related behavior. Physiol Behav 2024; 279:114533. [PMID: 38552707 DOI: 10.1016/j.physbeh.2024.114533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/26/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Increasing evidence suggests that the pre-conception parental environment has long-term consequences for offspring health and disease susceptibility. Though much of the work in this field concentrates on maternal influences, there is growing understanding that fathers also play a significant role in affecting offspring phenotypes. In this study, we investigate effects of altering the proportion of dietary fats and carbohydrates on paternal and offspring body composition and anxiety-related behavior in C57Bl/6-JArc mice. We show that in an isocaloric context, greater dietary fat increased body fat and reduced anxiety-like behavior of studs, whereas increased dietary sucrose had no significant effect. These dietary effects were not reflected in offspring traits, rather, we found sex-specific effects that differed between offspring body composition and behavioral traits. This finding is consistent with past paternal effect studies, where transgenerational effects have been shown to be more prominent in one sex over the other. Here, male offspring of fathers fed high-fat diets were heavier at 10 weeks of age due to increased lean body mass, whereas paternal diet had no significant effect on female offspring body fat or lean mass. In contrast, paternal dietary sugar appeared to have the strongest effects on male offspring behavior, with male offspring of high-sucrose fathers spending less time in the closed arms of the elevated plus maze. Both high-fat and high-sugar paternal diets were found to reduce anxiety-like behavior of female offspring, although this effect was only evident when offspring were fed a control diet. This study provides new understanding of the ways in which diet can shape the behavior of fathers and their offspring and contribute to the development of dietary guidelines to improve obesity and mental health conditions, such as anxiety.
Collapse
Affiliation(s)
- Therese Freire
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney NSW, Australia.
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - Flora Mackay
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Angela J Crean
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| |
Collapse
|
23
|
Broséus L, Guilbert A, Hough I, Kloog I, Chauvaud A, Seyve E, Vaiman D, Heude B, Chevrier C, Tost J, Slama R, Lepeule J. Placental DNA methylation signatures of prenatal air pollution exposure and potential effects on birth outcomes: an analysis of three prospective cohorts. Lancet Planet Health 2024; 8:e297-e308. [PMID: 38723642 DOI: 10.1016/s2542-5196(24)00045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/16/2024] [Accepted: 03/20/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Pregnancy air pollution exposure (PAPE) has been linked to a wide range of adverse birth and childhood outcomes, but there is a paucity of data on its influence on the placental epigenome, which can regulate the programming of physiological functions and affect child development. This study aimed to investigate the association between prenatal air pollutant exposure concentrations and changes in placental DNA methylation patterns, and to explore the potential windows of susceptibility and sex-specific alterations. METHODS This multi-site study used three prospective population-based mother-child cohorts: EDEN, PELAGIE, and SEPAGES, originating from four French geographical regions (Nancy, Poitiers, Brittany, and Grenoble). Pregnant women were included between 2003 and 2006 for EDEN and PELAGIE, and between 2014 and 2017 for SEPAGES. The main eligibility criteria were: being older than 18 years, having a singleton pregnancy, and living and planning to deliver in one of the maternity clinics in one of the study areas. A total of 1539 mother-child pairs were analysed, measuring placental DNA methylation using Illumina BeadChips. We used validated spatiotemporally resolved models to estimate PM2·5, PM10, and NO2 exposure over each trimester of pregnancy at the maternal residential address. We conducted a pooled adjusted epigenome-wide association study to identify differentially methylated 5'-C-phosphate-G-3' (CpG) sites and regions (assessed using the Infinium HumanMethylationEPIC BeadChip array, n=871), including sex-specific and sex-linked alterations, and independently validated our results (assessed using the Infinium HumanMethylation450 BeadChip array, n=668). FINDINGS We identified four CpGs and 28 regions associated with PAPE in the total population, 469 CpGs and 87 regions in male infants, and 150 CpGs and 66 regions in female infants. We validated 35% of the CpGs available. More than 30% of the identified CpGs were related to one (or more) birth outcome and most significant alterations were enriched for neural development, immunity, and metabolism related genes. The 28 regions identified for both sexes overlapped with imprinted genes (four genes), and were associated with neurodevelopment (nine genes), immune system (seven genes), and metabolism (five genes). Most associations were observed for the third trimester for female infants (134 of 150 CpGs), and throughout pregnancy (281 of 469 CpGs) and the first trimester (237 of 469 CpGs) for male infants. INTERPRETATION These findings highlight the molecular pathways through which PAPE might affect child health in a widespread and sex-specific manner, identifying the genes involved in the major physiological functions of a developing child. Further studies are needed to elucidate whether these epigenetic changes persist and affect health later in life. FUNDING French Agency for National Research, Fondation pour la Recherche Médicale, Fondation de France, and the Plan Cancer.
Collapse
Affiliation(s)
- Lucile Broséus
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France.
| | - Ariane Guilbert
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Ian Hough
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France; Institute of Environmental Geosciences, Université Grenoble Alpes, Grenoble, France; Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Be'er Sheva, Israel; Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anath Chauvaud
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Emie Seyve
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Daniel Vaiman
- Institut Cochin, U1016 Inserm, Unité Mixte de Recherche 8104, CNRS, Paris-Descartes University, Paris, France
| | - Barbara Heude
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, INRAE, Centre de Recherche en Épidémiologie et Statistiques, Paris, France
| | - Cécile Chevrier
- University of Rennes, Inserm, Ecole des Hautes Etudes en Santé Publique, Institut de Recherche en Santé, Environnement et Travail, Unité Mixte de Recherche 1085, Rennes, France
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut de Biologie François Jacob, University Paris Saclay, Evry, France
| | - Rémy Slama
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Johanna Lepeule
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology applied to Development and Respiratory Health, IAB, Grenoble, France.
| |
Collapse
|
24
|
Wang YQ, Zhang Y, Tang WF, Luo ZC, Zhang YT, Yan CH, Zhang J, Chen Q. Environmental antibiotics exposure and childhood obesity: A cross-sectional case-control study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116339. [PMID: 38669873 DOI: 10.1016/j.ecoenv.2024.116339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024]
Abstract
Children's exposures to environmental antibiotics are a major public health concern. However, limited data are available on the effects of environmental antibiotic exposures on childhood obesity. Our study aimed to explore this relationship. We conducted a cross-sectional case-control study nested in a population-based survey of primary school students, including 1855 obese and 1875 random selected control children. A total of 10 antibiotics in urine samples were measured by liquid chromatography-tandem mass spectrometry. Multivariable survey logistic regression was used to assess the associations between environmental antibiotics exposures and childhood obesity. After adjusting for potential confounders, increased odds of obesity were observed in children exposed to tetracycline (OR = 1.31, 95% CI: 1.09-1.57) and sulfamonomethoxine (OR = 1.43, 95% CI: 1-2.05). Comparing none (
Collapse
Affiliation(s)
- Yu-Qing Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Wei-Feng Tang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong-Cheng Luo
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Obstetrics and Gynecology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Faculty of Medicine, and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
| | - Yun-Ting Zhang
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chong-Huai Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qian Chen
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Czamara D, Dieckmann L, Lahti-Pulkkinen M, Cruceanu C, Henrich W, Plagemann A, Räikkönen K, Braun T, Binder EB, Lahti J, Entringer S. Sex differences in DNA methylation across gestation: a large scale, cross-cohort, multi-tissue analysis. Cell Mol Life Sci 2024; 81:177. [PMID: 38600394 PMCID: PMC11006734 DOI: 10.1007/s00018-024-05208-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024]
Abstract
Biological sex is a key variable influencing many physiological systems. Disease prevalence as well as treatment success can be modified by sex. Differences emerge already early in life and include pregnancy complications and adverse birth outcomes. The placenta is a critical organ for fetal development and shows sex-based differences in the expression of hormones and cytokines. Epigenetic regulation, such as DNA methylation (DNAm), may underlie the previously reported placental sexual dimorphism. We associated placental DNAm with fetal sex in three cohorts. Individual cohort results were meta-analyzed with random-effects modelling. CpG-sites differentially methylated with sex were further investigated regarding pathway enrichment, overlap with methylation quantitative trait loci (meQTLs), and hits from phenome-wide association studies (PheWAS). We evaluated the consistency of findings across tissues (CVS, i.e. chorionic villus sampling from early placenta, and cord blood) as well as with gene expression. We identified 10,320 epigenome-wide significant sex-differentially methylated probes (DMPs) spread throughout the epigenome of the placenta at birth. Most DMPs presented with lower DNAm levels in females. DMPs mapped to genes upregulated in brain, were enriched for neurodevelopmental pathways and significantly overlapped with meQTLs and PheWAS hits. Effect sizes were moderately correlated between CVS and placenta at birth, but only weakly correlated between birth placenta and cord blood. Sex differential gene expression in birth placenta was less pronounced and implicated genetic regions only marginally overlapped with those associated with differential DNAm. Our study provides an integrative perspective on sex-differential DNAm in perinatal tissues underscoring the possible link between placenta and brain.
Collapse
Affiliation(s)
- Darina Czamara
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Linda Dieckmann
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cristiana Cruceanu
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Wolfgang Henrich
- Department of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Andreas Plagemann
- Department of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, HUS Helsinki University Hospital, Helsinki, Finland
| | - Thorsten Braun
- Department of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sonja Entringer
- Institute of Medical Psychology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany.
- Department of Pediatrics, Health and Disease Research Program, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
26
|
Gan H, Xing Y, Tong J, Lu M, Yan S, Huang K, Wu X, Tao S, Gao H, Pan Y, Dai J, Tao F. Impact of Gestational Exposure to Individual and Combined Per- and Polyfluoroalkyl Substances on a Placental Structure and Efficiency: Findings from the Ma'anshan Birth Cohort. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:6117-6127. [PMID: 38525964 DOI: 10.1021/acs.est.3c09611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Prenatal exposure to perfluoroalkyl and polyfluoroalkyl substances (PFASs) is inevitable among pregnant women. Nevertheless, there is a scarcity of research investigating the connections between prenatal PFAS exposure and the placental structure and efficiency. Based on 712 maternal-fetal dyads in the Ma'anshan Birth Cohort, we analyzed associations between individual and mixed PFAS exposure and placental measures. We repeatedly measured 12 PFAS in the maternal serum during pregnancy. Placental weight, scaling exponent, chorionic disc area, and disc eccentricity were used as the outcome variables. Upon adjusting for confounders and implementing corrections for multiple comparisons, we identified positive associations between branched perfluorohexane sulfonate (br-PFHxS) and 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA) with placental weight. Additionally, a positive association was observed between br-PFHxS and the scaling exponent, where a higher scaling exponent signified reduced placental efficiency. Based on neonatal sex stratification, female infants were found to be more susceptible to the adverse effects of PFAS exposure. Mixed exposure modeling revealed that mixed PFAS exposure was positively associated with placental weight and scaling exponent, particularly during the second and third trimesters. Furthermore, br-PFHxS and 6:2 Cl-PFESA played major roles in the placental measures. This study provides the first epidemiological evidence of the relationship between prenatal PFAS exposure and placental measures.
Collapse
Affiliation(s)
- Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Yanan Xing
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Mengjuan Lu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan 243011 Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Shuman Tao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hui Gao
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022 Anhui, China
| | - Yitao Pan
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| |
Collapse
|
27
|
Griffith JA, King RD, Dunn AC, Lewis SE, Maxwell BA, Nurkiewicz TR, Goldsmith WT, Kelley EE, Bowdridge EC. Maternal nano-titanium dioxide inhalation exposure alters placental cyclooxygenase and oxidant balance in a sexually dimorphic manner. ADVANCES IN REDOX RESEARCH 2024; 10:10.1016/j.arres.2023.100090. [PMID: 38562524 PMCID: PMC10979698 DOI: 10.1016/j.arres.2023.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The placenta plays a critical role in nutrient-waste exchange between the maternal and fetal circulation, and thus impacts fetal growth and development. We have previously shown that nano-titanium dioxide (nano-TiO2) inhalation exposure during gestation decreased fetal female pup and placenta mass [1], which persists in the following generation [2]. In utero exposed females, once mated, their offspring's placentas had increased capacity for H2O2 production. Generation of oxidants such as hydrogen peroxide (H2O2), have been shown to impact cyclooxygenase activity, specifically metabolites such as prostacyclin (PGI2) or thromboxane (TXA2). Therefore, we hypothesized that maternal nano-TiO2 inhalation exposure during gestation results in alterations in placental production of prostacyclin and thromboxane mediated by enhanced H2O2 production in a sexually dimorphic manner. Pregnant Sprague-Dawley rats were exposed to nano-TiO2 aerosols or filtered air (sham--control) from gestational day (GD) 10-19. Dams were euthanized on GD 20, and fetal serum and placental tissue were collected based on fetal sex. Fetal placental zones (junctional zone (JZ) and labyrinth zone (LZ)) were assessed for xanthine oxidoreductase (XOR) activity, H2O2, and catalase activity, as well as 6-keto-PGF1α and TXB2 levels. Nano-TiO2 exposed fetal female LZ demonstrated significantly greater XOR activity compared to exposed males. Exposed fetal female LZ also demonstrated significantly diminished catalase activity compared to sham-control females. Exposed fetal female LZ had significantly increased abundance of 6-keto-PGF1α compared to sham-control females and increased TXB2 compared to exposed males. In the aggregate these data indicate that maternal nano-TiO2 inhalation exposure has a greater impact on redox homeostasis and PGI2/TXA2 balance in the fetal female LZ. Future studies need to address if treatment with an XO inhibitor during gestation can prevent diminished fetal female growth during maternal nano-TiO2 inhalation exposure.
Collapse
Affiliation(s)
- Julie A. Griffith
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Rachel D. King
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Allison C. Dunn
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sara E. Lewis
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brooke A. Maxwell
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R. Nurkiewicz
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T. Goldsmith
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Eric E. Kelley
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth C. Bowdridge
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
28
|
Scarborough J, Iachizzi M, Schalbetter SM, Müller FS, Weber-Stadlbauer U, Richetto J. Prenatal and postnatal influences on behavioral development in a mouse model of preconceptional stress. Neurobiol Stress 2024; 29:100614. [PMID: 38357099 PMCID: PMC10865047 DOI: 10.1016/j.ynstr.2024.100614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/12/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024] Open
Abstract
Depression during pregnancy is detrimental for the wellbeing of the expectant mother and can exert long-term consequences on the offspring's development and mental health. In this context, both the gestational environment and the postpartum milieu may be negatively affected by the depressive pathology. It is, however, challenging to assess whether the contributions of prenatal and postnatal depression exposure are distinct, interactive, or cumulative, as it is unclear whether antenatal effects are due to direct effects on fetal development or because antenatal symptoms continue postnatally. Preclinical models have sought to answer this question by implementing stressors that induce a depressive-like state in the dams during pregnancy and studying the effects on the offspring. The aim of our present study was to disentangle the contribution of direct stress in utero from possible changes in maternal behavior in a novel model of preconceptional stress based on social isolation rearing (SIR). Using a cross-fostering paradigm in this model, we show that while SIR leads to subtle changes in maternal behavior, the behavioral changes observed in the offspring are driven by a complex interaction between sex, and prenatal and postnatal maternal factors. Indeed, male offspring are more sensitive to the prenatal environment, as demonstrated by behavioral and transcriptional changes driven by their birth mother, while females are likely affected by more complex interactions between the pre and the postpartum milieu, as suggested by the important impact of their surrogate foster mother. Taken together, our findings suggest that male and female offspring have different time-windows and behavioral domains of susceptibility to maternal preconceptional stress, and thus underscore the importance of including both sexes when investigating the mechanisms that mediate the negative consequences of exposure to such stressor.
Collapse
Affiliation(s)
- Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Monica Iachizzi
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Sina M. Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Flavia S. Müller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Qiu F, Zhang H, Cui Y, Zhang L, Zhou W, Huang M, Xia W, Xu S, Li Y. Associations of maternal urinary rare earth elements individually and in mixtures with neonatal size at birth. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 343:123163. [PMID: 38104763 DOI: 10.1016/j.envpol.2023.123163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/21/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Prenatal rare earth elements (REEs) exposure is linked to unfavorable health consequences. Epidemiologic research on repeated measurements of REEs during gestation correlated with fetal growth is exiguous. Until now, few studies have characterized exposure characteristics of REEs in pregnant women. We aimed to ascertain the characteristics and predictors of REEs exposure over three trimesters among pregnant women and examine the possible effects of prenatal REEs exposure on size at birth. Urinary REEs concentrations exhibited considerable within-subject variation with intraclass correlation coefficients ranging from 0.16 to 0.58. Maternal age, household income, gestational weight gain, passive smoking during pregnancy, parity, and neonatal gender were associated with maternal urinary REEs concentrations. Elevated maternal urinary holmium and thulium concentrations in the 3rd trimester were significantly related to reductions in birth weight. Weighted quantile sum (WQS) regression model identified that urinary REEs mixture in the 3rd trimester were negatively related to birth weight (WQSREEs β = -26.22; 95% confidence interval [CI]: -47.62, -4.82), with holmium (40%) and thulium (24%) receiving the highest weights. Male infants received the most weight (>50%) related to decreased birth weight. This study revealed a significant association between individual and mixture REE exposure in late pregnancy with a reduction in birth weight.
Collapse
Affiliation(s)
- Feng Qiu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hongling Zhang
- Wuchang University of Technology, Wuhan, Hubei, People's Republic of China
| | - Yuan Cui
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Liping Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wensi Zhou
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Min Huang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
30
|
Zhao M, Lei J, Deng F, Zhao C, Xu T, Ji B, Fu M, Wang X, Sun M, Zhang M, Gao Q. Gestational Hypoxia Impaired Endothelial Nitric Oxide Synthesis Via miR-155-5p/NADPH Oxidase/Reactive Oxygen Species Axis in Male Offspring Vessels. J Am Heart Assoc 2024; 13:e032079. [PMID: 38240225 PMCID: PMC11056123 DOI: 10.1161/jaha.123.032079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Nitric oxide (NO) is the most important vasodilator secreted by vascular endothelial cells, and its abnormal synthesis is involved in the development of cardiovascular disease. The prenatal period is a critical time for development and largely determines lifelong vascular health in offspring. Given the high incidence and severity of gestational hypoxia in mid-late pregnancy, it is urgent to further explore whether it affects the long-term synthesis of NO in offspring vascular endothelial cells. METHODS AND RESULTS Pregnant Sprague-Dawley rats were housed in a normoxic or hypoxic (10.5% O2) chamber from gestation days 10 to 20. The thoracic aortas of fetal and adult male offspring were isolated for experiments. Gestational hypoxia significantly reduces the NO-dependent vasodilation mediated by acetylcholine in both the fetal and adult offspring thoracic aorta rings. Meanwhile, acetylcholine-induced NO synthesis is impaired in vascular endothelial cells from hypoxic offspring thoracic aortas. We demonstrate that gestational hypoxic offspring exhibit a reduced endothelial NO synthesis capacity, primarily due to increased expression of NADPH oxidase 2 and enhanced reactive oxygen species. Additionally, gestational hypoxic offspring show elevated levels of miR-155-5p in vascular endothelial cells, which is associated with increased expression of NADPH oxidase 2 and reactive oxygen species generation, as well as impaired NO synthesis. CONCLUSIONS The present study is the first to demonstrate that gestational hypoxia impairs endothelial NO synthesis via the miR-155-5p/NADPH oxidase 2/reactive oxygen species axis in offspring vessels. These novel findings indicate that the detrimental effects of gestational hypoxia on fetal vascular function can persist into adulthood, providing new insights into the development of vascular diseases.
Collapse
Affiliation(s)
- Meng Zhao
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of Obstetrics and GynecologyThe Third People’s Hospital of Bengbu Affiliated to Bengbu Medical CollegeBengbuAnhui ProvinceChina
| | - Jiahui Lei
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fengying Deng
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chenxuan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Bingyu Ji
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mengyu Fu
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xietong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
| | - Miao Sun
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health CommissionShandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao UniversityJinanShandongChina
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
| | - Qinqin Gao
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
31
|
Hercus JC, Metcalfe KX, Christians JK. Sex differences in growth and mortality in pregnancy-associated hypertension. PLoS One 2024; 19:e0296853. [PMID: 38206980 PMCID: PMC10783718 DOI: 10.1371/journal.pone.0296853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND It is hypothesized that male fetuses prioritize growth, resulting in increased mortality, whereas females reduce growth in the presence of adversity. Preeclampsia reflects a chronic condition, in which fetuses have the opportunity to adjust growth. If females reduce their growth in response to preeclampsia, but males attempt to maintain growth at the cost of survival, we predict that differences in birthweight between preeclamptic and non-preeclamptic pregnancies will be greater among females, whereas differences in mortality will be greater among males. METHODS We analysed data from the Centers for Disease Control and Prevention. We compared pregnancies with pregnancy-associated hypertension (PAH) and controls. RESULTS The difference in birthweight between pregnancies affected by PAH and controls varied by fetal sex and gestational age. Among pregnancies of White individuals, at 34-35 weeks, the difference between PAH and controls was higher among females, as predicted. However, this pattern was reversed earlier in pregnancy and around term. Such variation was not significant in Black pregnancies. In both Black and White pregnancies, early in gestation, males had lower odds of death in PAH pregnancies, but higher odds of death in control pregnancies, counter to our prediction. Later, males had higher odds of death in PAH and controls, although the increased odds of death in males was not higher in PAH pregnancies than in controls. Overall, the difference in birthweight between surviving and non-surviving infants was greater in males than in females, opposite to our prediction. CONCLUSIONS The impact of PAH on birthweight and survival varies widely throughout gestation. Differences in birthweight and survival between male and female PAH and controls are generally not consistent with the hypothesis that males prioritize fetal growth more than females, and that this is a cause of increased mortality in males.
Collapse
Affiliation(s)
- Jess C. Hercus
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Katherine X. Metcalfe
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian K. Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, 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, British Columbia, Canada
| |
Collapse
|
32
|
Ryva BA, Pacyga DC, Anderson KY, Calafat AM, Whalen J, Aung MT, Gardiner JC, Braun JM, Schantz SL, Strakovsky RS. Associations of urinary non-persistent endocrine disrupting chemical biomarkers with early-to-mid pregnancy plasma sex-steroid and thyroid hormones. ENVIRONMENT INTERNATIONAL 2024; 183:108433. [PMID: 38219543 PMCID: PMC10858740 DOI: 10.1016/j.envint.2024.108433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/22/2023] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND/OBJECTIVES Pregnant women are exposed to numerous endocrine disrupting chemicals (EDCs) that can affect hormonal pathways regulating pregnancy outcomes and fetal development. Thus, we evaluated overall and fetal sex-specific associations of phthalate/replacement, paraben, and phenol biomarkers with sex-steroid and thyroid hormones. METHODS Illinois women (n = 302) provided plasma for progesterone, estradiol, testosterone, free T4 (FT4), total T4 (TT4), and thyroid stimulating hormone (TSH) at median 17 weeks gestation. Women also provided up-to-five first-morning urine samples monthly across pregnancy (8-40 weeks), which we pooled to measure 19 phthalate/replacement metabolites (reflecting ten parent compounds), three parabens, and six phenols. We used linear regression to evaluate overall and fetal sex-specific associations of biomarkers with hormones, as well as weighted quantile sum and Bayesian kernel machine regression (BKMR) to assess cumulative associations, non-linearities, and chemical interactions. RESULTS In women of relatively high socioeconomic status, several EDC biomarkers were associated with select hormones, without cumulative or non-linear associations with progesterone, FT4, or TT4. The biomarker mixture was negatively associated with estradiol (only at higher biomarker concentrations using BKMR), testosterone, and TSH, where each 10% mixture increase was associated with -5.65% (95% CI: -9.79, -1.28) lower testosterone and -0.09 μIU/mL (95% CI: -0.20, 0.00) lower TSH. Associations with progesterone, testosterone, and FT4 did not differ by fetal sex. However, in women carrying females, we identified an inverted u-shaped relationship of the mixture with estradiol. Additionally, in women carrying females, each 10% increase in the mixture was associated with 1.50% (95% CI: -0.15, 3.18) higher TT4, whereas in women carrying males, the mixture was associated with -1.77% (95% CI: -4.08, 0.58) lower TT4 and -0.18 μIU/mL (95% CI: -0.33, -0.03) lower TSH. We also identified select chemical interactions. CONCLUSION Some biomarkers were associated with early-to-mid pregnancy hormones. There were some sex-specific and non-linear associations. Future studies could consider how these findings relate to pregnancy/birth outcomes.
Collapse
Affiliation(s)
- Brad A Ryva
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, United States; College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Diana C Pacyga
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States; Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States
| | - Kaitlyn Y Anderson
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, United States
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, United States
| | - Jason Whalen
- Michigan Diabetes Research Center Chemistry Laboratory, University of Michigan, Ann Arbor, MI 48109, United States
| | - Max T Aung
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, United States
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI 02912, United States
| | - Susan L Schantz
- The Beckman Institute, University of Illinois, Urbana-Champaign, IL 61801, United States; Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61802, United States
| | - Rita S Strakovsky
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States; Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
33
|
Wang H, Zhang L, Wu J, Wang P, Li Q, Sui X, Xu Y, Zhao Y, Liu Y, Zhang Y. Sex-specific effects of organophosphate ester exposure on child growth trajectories in the first two years. ECO-ENVIRONMENT & HEALTH 2023; 2:152-160. [PMCID: PMC10702896 DOI: 10.1016/j.eehl.2023.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 12/10/2023]
Abstract
The connections between urinary organophosphate ester (OPE) metabolites and child growth have been identified in prior research, but there is currently a dearth of epidemiological evidence regarding the sex-specific impact of OPEs on child growth trajectories. This study enrolled 804 maternal–child pairs, and five OPE congeners were quantified in maternal serum during pregnancy. In this study, the impact of prenatal OPE exposure on child growth trajectories was assessed using linear mixed-effect models and a group-based trajectory model (GBTM), with consideration given to sex-specific effects. Fetuses were frequently exposed to OPEs in utero, and tris(2-butoxyethel) phosphate (TBEP) exhibited the highest concentration levels in maternal serum. Among male children, an increase of 2.72 ng/g lipid in TBEP concentration was associated with a 0.11-unit increase in head circumference-for-age z-score (HCAZ), and the effect was mainly concentrated at 1 and 2 months of age. Among female children, an increase of 2.72 ng/g lipid in tris(2-chloro-1-(chloromethyl) ethyl) phosphate (TDCPP) concentration was associated with a 0.15-unit increase in length-for-age z-score (LAZ) and a 0.14-unit increase in weight-for-age z-score (WAZ), and the effects were mainly concentrated at 9 months of age. For HCAZ trajectories, higher prenatal TBEP exposure was associated with higher odds for the fast growth group in male children. For the LAZ and WAZ trajectories, higher prenatal TDCPP exposure was associated with higher odds for the fast growth group in female children. The trajectory analysis approach provided insight into the complex associations between OPE exposure and child growth. •Organophosphate ester (OPEs) were detected in serum samples of pregnant women, and tributyl phosphate (TBP) was the most frequently detected OPEs. •Prenatal exposure to TBP, tris(2-butoxyethyl) phosphate (TBEP), and tris(2-chloro-1-(chloromethyl) ethyl) phosphate (TDCPP) were positively associated with infant growth trajectories. •Female infants were more sensitive to OPE exposure than males.
Collapse
Affiliation(s)
- Hang Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Liyi Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jie Wu
- The Maternal and Child Healthcare Hospital of Songjiang District, Shanghai 201600, China
| | - Pengpeng Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Qiang Li
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xinyao Sui
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yaqi Xu
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yue Zhao
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yang Liu
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yunhui Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China (Fudan University), Shanghai 200032, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Sundrani DP, Joshi SR. Assisted reproductive technology (ART) and epigenetic modifications in the placenta. HUM FERTIL 2023; 26:665-677. [PMID: 34706609 DOI: 10.1080/14647273.2021.1995901] [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: 07/16/2020] [Accepted: 04/19/2021] [Indexed: 10/20/2022]
Abstract
Assisted reproductive technology (ART) has become common amongst couples with infertility issues. ART is known to be successful, but epidemiological data indicates that ART is associated with placental disorders. Additionally, reports show increased risks of short- and long-term complications in children born to mothers undergoing ART. However, the mechanisms responsible for these events are obscure. The placenta is considered as a key organ for programming of diseases and ART procedures are suggested to alter the placental function and intrauterine growth trajectories. Epigenetic changes in maternal and foetal tissues are suggested to be the underlying mechanisms for these outcomes. Epigenetic regulation is known to evolve following fertilisation and before implantation and subsequently across gestation. During these critical periods of epigenetic 'programming', DNA methylation and chromatin remodelling influence the placental structure and function by regulating the expression of various genes. ART treatment coinciding with epigenetic 'programming' events during gametogenesis and early embryo development may alter the programming phases leading to long-term consequences. Thus, disruptions in placental development observed in ART pregnancies could be associated with altered epigenetic regulation of vital genes in the placenta. The review summarises available literature on the influence of ART procedures on epigenetic changes in the placenta.
Collapse
Affiliation(s)
- Deepali P Sundrani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|
35
|
Stenhouse C, Bazer FW, Ashworth CJ. Sexual dimorphism in placental development and function: Comparative physiology with an emphasis on the pig. Mol Reprod Dev 2023; 90:684-696. [PMID: 35466463 DOI: 10.1002/mrd.23573] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022]
Abstract
Across mammalian species, it has been demonstrated that sex influences birth weight, with males being heavier than females; a characteristic that can be observed from early gestation. Male piglets are more likely to be stillborn and have greater preweaning mortality than their female littermates, despite the additional maternal investment into male fetal growth. Given the conserved nature of the genome between the sexes, it is hypothesized that these developmental differences between males and females are most likely orchestrated by differential placental adaptation. This review summarizes the current understanding of fetal sex-specific differences in placental and endometrial structure and function, with an emphasis on pathways found to be differentially regulated in the pig including angiogenesis, apoptosis, and proliferation. Given the importance of piglet sex in agricultural enterprises, and the potential for skewed litter sex ratios, it is imperative to improve understanding of the relationship between fetal sex and molecular signaling in both the placenta and endometria across gestation.
Collapse
Affiliation(s)
- Claire Stenhouse
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Cheryl J Ashworth
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
36
|
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
|
37
|
Wu Y, Zeng F, Li J, Jiang Y, Zhao S, Knibbs LD, Zhang X, Wang Y, Zhang Q, Wang Q, Hu Q, Guo X, Chen Y, Cao G, Wang J, Yang X, Wang X, Liu T, Zhang B. Sex-specific relationships between prenatal exposure to metal mixtures and birth weight in a Chinese birth cohort. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115158. [PMID: 37348214 DOI: 10.1016/j.ecoenv.2023.115158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
Birth weight is an indicator linking intrauterine environmental exposures to later-life diseases, and intrauterine metal exposure may affect birth weight in a sex-specific manner. We investigated sex-specific associations between prenatal exposure to metal mixtures and birth weight in a Chinese birth cohort. The birth weight of 1296 boys and 1098 girls were recorded, and 10 metals in maternal urine samples collected during pregnancy were measured using inductively coupled plasma mass spectrometry. Bayesian Kernel Machine Regression was used to estimate the association of individual metals or metal mixtures and birth weight for gestational age (BW for GA). The model showed a sex-specific relationship between prenatal exposure to metal mixtures and BW for GA with a significant negative association in girls and a non-significant positive association in boys. Cadmium (Cd) and nickel (Ni) were positively and negatively associated with BW for GA in girls, respectively. Moreover, increasing thallium (Tl) concentration lowered the positive association between Cd and BW for GA and enhanced the negative association between Ni and BW for GA in girls. When exposure to other metals increased, the positive association with Cd diminished, whereas the negative association with Ni or Tl increased. Our findings provide evidence supporting the complex effects of intrauterine exposure to metal mixtures on the birth weight of girls and further highlight the sex heterogeneity in fetal development influenced by intrauterine environmental factors.
Collapse
Affiliation(s)
- Ying Wu
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fulin Zeng
- Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinhui Li
- Department of Urology, Stanford University Medical Center, Stanford, CA, USA
| | - Yukang Jiang
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern China Center for Statistical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China; Chinese University of Hong Kong (CUHK) Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Luke D Knibbs
- School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Xiaojun Zhang
- Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiding Wang
- Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Qianqian Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiong Wang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiansheng Hu
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaobo Guo
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern China Center for Statistical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yumeng Chen
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Ganxiang Cao
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jing Wang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Xingfen Yang
- Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Xueqin Wang
- Department of Statistics and Finance/International Institute of Finance, School of Management, University of Science and Technology of China, Hefei, Anhui, China
| | - Tao Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Disease Control and Prevention Institute of Jinan University, Jinan University, Guangzhou, Guangdong, China.
| | - Bo Zhang
- Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Paz-Sabillón M, Torres-Sánchez L, Piña-Pozas M, Del Razo LM, Quintanilla-Vega B. Prenatal Exposure to Potentially Toxic Metals and Their Effects on Genetic Material in Offspring: a Systematic Review. Biol Trace Elem Res 2023; 201:2125-2150. [PMID: 35713810 DOI: 10.1007/s12011-022-03323-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022]
Abstract
In recent years, the background level of environmental pollutants, including metals, has increased. Pollutant exposure during the earliest stages of life may determine chronic disease susceptibility in adulthood because of genetic or epigenetic changes. The objective of this review was to identify the association between prenatal and early postnatal exposure to potentially toxic metals (PTMs) and their adverse effects on the genetic material of offspring. A systematic review was carried out following the Cochrane methodology in four databases: PubMed, Scopus, Web of Science, and the Cochrane Library. Eligible papers were those conducted in humans and published in English between 2010/01/01 and 2021/04/30. A total of 57 articles were included, most of which evaluated prenatal exposure. Most commonly evaluated PTMs were As, Cd, and Pb. Main adverse effects on the genetic material of newborns associated with PTM prenatal exposure were alterations in telomere length, gene or protein expression, mitochondrial DNA content, metabolomics, DNA damage, and epigenetic modifications. Many of these effects were sex-specific, being predominant in boys. One article reported a synergistic interaction between As and Hg, and two articles observed antagonistic interactions between PTMs and essential metals, such as Cu, Se, and Zn. The findings in this review highlight that the problem of PTM exposure persists, affecting the most susceptible populations, such as newborns. Some of these associations were observed at low concentrations of PTMs. Most of the studies have focused on single exposures; however, three interactions between essential and nonessential metals were observed, highlighting that metal mixtures need more attention.
Collapse
Affiliation(s)
- Marvin Paz-Sabillón
- Department of Toxicology, Cinvestav, Ave. IPN 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - Luisa Torres-Sánchez
- National Institute of Public Health, Ave. Universidad 655, Santa María Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico
| | - Maricela Piña-Pozas
- National Institute of Public Health, Ave. Universidad 655, Santa María Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico
| | - Luz M Del Razo
- Department of Toxicology, Cinvestav, Ave. IPN 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - Betzabet Quintanilla-Vega
- Department of Toxicology, Cinvestav, Ave. IPN 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico.
| |
Collapse
|
39
|
Maxwell A, Adzibolosu N, Hu A, You Y, Stemmer PM, Ruden DM, Petriello MC, Sadagurski M, Debarba LK, Koshko L, Ramadoss J, Nguyen AT, Richards D, Liao A, Mor G, Ding J. Intrinsic sexual dimorphism in the placenta determines the differential response to benzene exposure. iScience 2023; 26:106287. [PMID: 37153445 PMCID: PMC10156617 DOI: 10.1016/j.isci.2023.106287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/09/2022] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Maternal immune activation (MIA) by environmental challenges is linked to severe developmental complications, such as neurocognitive disorders, autism, and even fetal/maternal death. Benzene is a major toxic compound in air pollution that affects the mother as well as the fetus and has been associated with reproductive complications. Our objective was to elucidate whether benzene exposure during gestation triggers MIA and its impact on fetal development. We report that benzene exposure during pregnancy leads MIA associated with increased fetal resorptions, fetal growth, and abnormal placenta development. Furthermore, we demonstrate the existence of a sexual dimorphic response to benzene exposure in male and female placentas. The sexual dimorphic response is a consequence of inherent differences between male and female placenta. These data provide crucial information on the origins or sexual dimorphism and how exposure to environmental factors can have a differential impact on the development of male and female offspring.
Collapse
Affiliation(s)
- Anthony Maxwell
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Nicholas Adzibolosu
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Anna Hu
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Yuan You
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Douglas M. Ruden
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Michael C. Petriello
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Lucas K. Debarba
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Lisa Koshko
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Jayanth Ramadoss
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | | | - Darby Richards
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Gil Mor
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Jiahui Ding
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
40
|
Wei Q, Zou J, Ma X, Xiao X, Zhang Y, Shi H. Prospective associations between various prenatal exposures to maternal psychological stress and neurodevelopment in children within 24 months after birth. J Affect Disord 2023; 327:101-110. [PMID: 36738998 DOI: 10.1016/j.jad.2023.01.103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND There is increasing evidence that prenatal exposure to maternal psychological distress may be a factor influencing offspring neurodevelopment, but stress type-dependent effects of maternal psychological distress on offspring neurodevelopment in early childhood have yet to be fully elucidated. Additionally, although positive maternal mental health exerts potential effects in protecting against adverse health outcomes, few investigators have considered the effects of positive maternal mental health on offspring neurodevelopment in early childhood. AIMS To determine the associations between various prenatal exposures to maternal psychological distress and positive life-event experiences and offspring neurodevelopment within 24 months of age. METHODS A total of 4412 mother-child dyads were recruited from the Shanghai Maternal-Child Pairs Cohort (Shanghai MCPC). Maternal perceived stress, negative life-event stress, positive life-event experiences around the time of conception (i.e., three months prior to and after conception) were assessed at 12-16 gestational weeks, and maternal anxiety and depressive symptoms were assessed at 32-36 gestational weeks. We measured children's neurodevelopment using the Ages and Stages Questionnaire, Third Edition (ASQ-3) at two, six, 12, and 24 months postnatally. We then exploited generalized linear models to estimate the associations between prenatal maternal psychological distress and positive life-event experiences and children's neurodevelopment at the above periods, and generalized linear mixed models were applied to assess the associations between maternal psychological distress and positive life-event experiences and suspected developmental delay (SDD) in children within 24 months after birth based on a longitudinal design. RESULTS Maternal perceived stress and negative life-event stress around the time of conception, and anxiety and depressive symptoms during late pregnancy were negatively associated with scores of children's neurodevelopment at two, six, 12, and 24 months of age; while maternal life-event experiences were positively associated with scores of children's neurodevelopment. Longitudinal analysis revealed that higher levels of maternal negative life-event stress and depressive symptoms augmented the risk of SDD in personal-social (OR = 1.435, 1.681). Mothers who experienced higher levels of positive life-event experiences exhibited a reduced risk of SDD in gross motor and personal-social domains (OR = 0.373, 0.350). CONCLUSIONS Prenatal exposure to maternal psychological distress is negatively associated with children's neurodevelopment in early childhood depending upon the type of distress. Maternal positive life-event experiences around the time of conception appeared to present potential benefits for child neurodevelopment.
Collapse
Affiliation(s)
- Qian Wei
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jiaojiao Zou
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xuemei Ma
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xirong Xiao
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Huijing Shi
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
41
|
Griffith JA, Dunn A, DeVallance E, Schafner KJ, Engles KJ, Batchelor TP, Goldsmith WT, Wix K, Hussain S, Bowdridge EC, Nurkiewicz TR. Maternal nano-titanium dioxide inhalation alters fetoplacental outcomes in a sexually dimorphic manner. FRONTIERS IN TOXICOLOGY 2023; 5:1096173. [PMID: 36950144 PMCID: PMC10025460 DOI: 10.3389/ftox.2023.1096173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
The placenta plays a critical role in nutrient-waste exchange between the maternal and fetal circulations, thus functioning as an interface that profoundly impacts fetal growth and development. The placenta has long been considered an asexual organ, but, due to its embryonic origin it shares the same sex as the fetus. Exposures to toxicant such as diesel exhaust, have been shown to result in sexually dimorphic outcomes like decreased placental mass in exposed females. Therefore, we hypothesize that maternal nano-TiO2 inhalation exposure during gestation alters placental hemodynamics in a sexually dimorphic manner. Pregnant Sprague-Dawley rats were exposed from gestational day 10-19 to nano-TiO2 aerosols (12.17 ± 1.69 mg/m3) or filtered air (sham-control). Dams were euthanized on GD20, and fetal tissue was collected based on fetal sex: whole placentas, placental junctional zone (JZ), and placental labyrinth zone (LZ). Fetal mass, placental mass, and placental zone percent areas were assessed for sex-based differences. Exposed fetal females were significantly smaller compared to their exposed male counterparts (2.65 ± 0.03 g vs 2.78 ± 0.04 g). Nano-TiO2 exposed fetal females had a significantly decreased percent junctional zone area compared to the sham-control females (24.37 ± 1.30% vs 30.39 ± 1.54%). The percent labyrinth zone area was significantly increased for nano-TiO2 females compared to sham-control females (75.63 ± 1.30% vs 69.61 ± 1.54%). Placental flow and hemodynamics were assessed with a variety of vasoactive substances. It was found that nano-TiO2 exposed fetal females only had a significant decrease in outflow pressure in the presence of the thromboxane (TXA2) mimetic, U46619, compared to sham-control fetal females (3.97 ± 1.30 mm Hg vs 9.10 ± 1.07 mm Hg) and nano-TiO2 fetal males (9.96 ± 0.66 mm Hg). Maternal nano-TiO2 inhalation exposure has a greater effect on fetal female mass, placental zone mass and area, and adversely impacts placental vasoreactivity. This may influence the female growth and development later in life, future studies need to further study the impact of maternal nano-TiO2 inhalation exposure on zone specific mechanisms.
Collapse
Affiliation(s)
- Julie A. Griffith
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Allison Dunn
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Evan DeVallance
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Kallie J. Schafner
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Kevin J. Engles
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Thomas P. Batchelor
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - William T. Goldsmith
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Kimberley Wix
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Salik Hussain
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Elizabeth C. Bowdridge
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Timothy R. Nurkiewicz
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, United States
| |
Collapse
|
42
|
Gan H, Zhang Y, Wang YF, Tao FB, Gao H. Relationships of prenatal organophosphate ester exposure with pregnancy and birth outcomes: A systematic scoping review of epidemiological studies. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114642. [PMID: 36791503 DOI: 10.1016/j.ecoenv.2023.114642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Exposure to organophosphate esters (OPEs) during pregnancy has been suggested to be associated with adverse pregnancy and birth outcomes. However, relevant investigations are scarce, and the findings are inconsistent. We aimed to conduct a scoping review to provide an overview of these associations. Electronic databases, including MEDLINE (through PubMed), Web of Science, and CNKI (China National Knowledge Infrastructure), were searched from inception to March 2022 and updated in July 2022. A total of 8 studies (1860 participants) were included. Limited evidence indicates that OPE exposure during pregnancy may be negatively associated with both maternal and neonatal triiodothyronine and tetraiodothyronine concentrations but positively associated with thyroid-stimulating hormone concentrations. OPE exposure during pregnancy may be associated with lower insulin concentrations. OPE exposure during pregnancy was associated with gestational age in a sex-specific manner. Intrauterine OPE exposure might increase the risk of preterm birth in female infants but decrease the risk of preterm birth in male infants. Prenatal OPE exposure might be associated with an increased risk of low birth weight. The current scoping review suggests that OPE exposure during pregnancy may disturb pregnancy and birth health, including adverse thyroid function and birth size. Because of the limited evidence obtained for most associations, additional studies followed by a traditional systematic review are needed to confirm these findings.
Collapse
Affiliation(s)
- Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China; Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Yi Zhang
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China; Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Ya-Fei Wang
- Nursing Department, Anhui Medical College, Hefei 230601, Anhui, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Hui Gao
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
43
|
Satokar VV, Derraik JGB, Harwood M, Okesene-Gafa K, Beck K, Cameron-Smith D, Garg ML, O'Sullivan JM, Sundborn G, Pundir S, Mason RP, Cutfield WS, Albert BB. Fish oil supplementation during pregnancy and postpartum in mothers with overweight and obesity to improve body composition and metabolic health during infancy: A double-blind randomized controlled trial. Am J Clin Nutr 2023; 117:883-895. [PMID: 36781129 DOI: 10.1016/j.ajcnut.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Maternal obesity during pregnancy is associated with an increased risk of obesity and metabolic disease in the offspring. Supplementation with fish oil (FO), which is insulin sensitizing, during pregnancy in mothers with overweight or obesity may prevent the development of greater adiposity and metabolic dysfunction in their children. OBJECTIVES To determine the effects of FO supplementation throughout the second half of pregnancy and lactation in mothers with overweight or obesity on infant body composition and metabolism. METHODS A double-blind randomized controlled trial of 6 g FO (3.55 g/d of n-3 PUFAs) compared with olive oil (control) from mid-pregnancy until 3 mo postpartum. Eligible women had singleton pregnancies at 12-20 wk of gestation, and BMI ≥ 25 kg/m2. The primary outcome was the infant body fat percentage (DXA scans) at 2 wk of age. Secondary outcomes included maternal metabolic markers during pregnancy, infant anthropometry at 2 wk and 3 mo of age, and metabolic markers at 3 mo. RESULTS A total of 129 mothers were randomized, and 98 infants had a DXA scan at 2 wk. PRIMARY OUTCOME Imputed and nonimputed analyses showed no effects of FO supplementation on infant body fat percentage at age 2 wk. SECONDARY OUTCOMES There were no treatment effects on infant outcomes at 2 wk, but FO infants had a higher BMI z-score (P = 0.025) and ponderal index (P = 0.017) at age 3 mo. FO supplementation lowered maternal triglycerides by 17% at 30 wk of pregnancy (P = 0.0002) and infant triglycerides by 21% at 3 mo of age (P = 0.016) but did not affect maternal or infant insulin resistance. The rate of emergency cesarean section was lower with FO supplementation [aRR = 0.38 (95%CI 0.16, 0.90); P = 0.027]. CONCLUSIONS FO supplementation of mothers with overweight or obesity during pregnancy did not impact infant body composition. There is a need to follow up the offspring to determine whether the observed metabolic effects persist. CLINICAL TRIAL REGISTRY NUMBER This study was registered with the Australian New Zealand Clinical Trials Registry (ACTRN12617001078347p). In addition, the Universal Trial Number, WHO, was obtained (U1111-1199-5860).
Collapse
Affiliation(s)
- Vidit V Satokar
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand; Department of Paediatrics: Child and Youth Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden; Environmental - Occupational Health Sciences and Non-Communicable Diseases Research Group, Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Matire Harwood
- Department of General Practice and Primary Care, University of Auckland, Auckland, New Zealand
| | - Karaponi Okesene-Gafa
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn Beck
- School of Sport Exercise and Nutrition, College of Health, Massey University, Auckland, New Zealand
| | - David Cameron-Smith
- Liggins Institute, University of Auckland, Auckland, New Zealand; College of Engineering, Science and Environment, University of Newcastle, New South Wales, Australia; School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, New South Wales, Australia
| | - Manohar L Garg
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, New South Wales, Australia
| | | | - Gerhard Sundborn
- Department of Pacific Health, School of Population Health, University of Auckland, Auckland, New Zealand
| | - Shikha Pundir
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - R Preston Mason
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand; A Better Start - National Science Challenge, University of Auckland, New Zealand
| | - Benjamin B Albert
- Liggins Institute, University of Auckland, Auckland, New Zealand; A Better Start - National Science Challenge, University of Auckland, New Zealand.
| |
Collapse
|
44
|
Ebbing C, Rasmussen S, Kessler J, Moster D. Association of placental and umbilical cord characteristics with cerebral palsy: national cohort study. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 61:224-230. [PMID: 36722428 PMCID: PMC10108292 DOI: 10.1002/uog.26047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 05/27/2023]
Abstract
OBJECTIVES Cerebral palsy (CP) is a group of movement disorders usually diagnosed in childhood. A substantial proportion are thought to be caused by antenatal events. Abnormalities of the umbilical cord and placenta are associated with an increased risk of adverse neonatal outcomes, but it is unclear whether these conditions also carry an increased risk of CP. We aimed to determine whether abnormalities of the umbilical cord or placenta are associated with CP and assess if these associations differ by sex of the child or gestational age at birth. METHODS We performed a national cohort study by linking data from The Medical Birth Registry of Norway with other national registries. All liveborn singletons born between 1999 and 2017 (n = 1 087 486) were included and followed up until the end of 2019. Diagnoses of CP were provided by the Norwegian National Insurance Scheme and the Norwegian Patient Register. We used generalized estimating equations and multilevel log binomial regression to calculate relative risks (RR), adjusted for year of birth, and stratified analyses were carried out based on sex and gestational age at birth. Exposures were abnormal umbilical cord (velamentous or marginal insertion, single umbilical artery (SUA), knots and entanglement), and placental abnormalities (retained placenta, placental abruption and previa). RESULTS A total of 2443 cases with CP (59.8% males) were identified. Velamentous cord insertion (adjusted RR (aRR), 2.11 (95% CI, 1.65-2.60)), cord knots (aRR, 1.53 (95% CI, 1.15-2.04)) and placental abnormalities (placenta previa (aRR, 3.03 (95% CI, 2.00-4.61)), placental abruption (aRR, 10.63 (95% CI, 8.57-13.18)) and retained placenta (aRR, 1.71 (95% CI, 1.32-2.22))) carried an increased risk of CP. Velamentous cord insertion was associated with CP regardless of gestational age or sex. A retained placenta was associated with a 2-fold increased risk for CP in males, while the associations of SUA and cord knot with CP were significant only among females. CONCLUSIONS The detection of placental and umbilical cord abnormalities may help identify children at increased risk of CP. The associations between placental or umbilical cord abnormalities and the risk of CP do not vary substantially with gestational age at birth or sex of the child. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- C. Ebbing
- Department of Obstetrics and GynecologyHaukeland University HospitalBergenNorway
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - S. Rasmussen
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - J. Kessler
- Department of Obstetrics and GynecologyHaukeland University HospitalBergenNorway
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - D. Moster
- Department of Global Public Health and Primary CareUniversity of BergenBergenNorway
- Department of PediatricsHaukeland University HospitalBergenNorway
| |
Collapse
|
45
|
Zengeler KE, Shapiro DA, Bruch KR, Lammert CR, Ennerfelt H, Lukens JR. SSRI treatment modifies the effects of maternal inflammation on in utero physiology and offspring neurobiology. Brain Behav Immun 2023; 108:80-97. [PMID: 36343752 PMCID: PMC10291741 DOI: 10.1016/j.bbi.2022.10.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/17/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Perturbations to the in utero environment can dramatically change the trajectory of offspring neurodevelopment. Insults commonly encountered in modern human life such as infection, toxins, high-fat diet, prescription medications, and others are increasingly linked to behavioral alterations in prenatally-exposed offspring. While appreciation is expanding for the potential consequence that these triggers can have on embryo development, there is a paucity of information concerning how the crucial maternal-fetal interface (MFI) responds to these various insults and how it may relate to changes in offspring neurodevelopment. Here, we found that the MFI responds both to an inflammatory state and altered serotonergic tone in pregnant mice. Maternal immune activation (MIA) triggered an acute inflammatory response in the MFI dominated by interferon signaling that came at the expense of ordinary development-related transcriptional programs. The major MFI compartments, the decidua and the placenta, each responded in distinct manners to MIA. MFIs exposed to MIA were also found to have disrupted sex-specific gene expression and heightened serotonin levels. We found that offspring exposed to MIA had sex-biased behavioral changes and that microglia were not transcriptionally impacted. Moreover, the combination of maternal inflammation in the presence of pharmacologic inhibition of serotonin reuptake further transformed MFI physiology and offspring neurobiology, impacting immune and serotonin signaling pathways alike. In all, these findings highlight the complexities of evaluating diverse environmental impacts on placental physiology and neurodevelopment.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Daniel A Shapiro
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine R Bruch
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Catherine R Lammert
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - Hannah Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
46
|
Bortolotto CC, dos Santos IDS, Vaz JDS, Matijasevich A. Gestational age, intrauterine growth and body composition at 11 years of age. Rev Saude Publica 2023; 56:116. [PMID: 36629707 PMCID: PMC9749740 DOI: 10.11606/s1518-8787.2022056004022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 01/28/2022] [Indexed: 12/28/2022] Open
Abstract
OBJETIVE To assess the association of gestational age (GA) and intrauterine growth with body composition at 11 years of age. METHOD Analysis of data from the 2004 Pelotas birth cohort, whose outcomes were fat mass (FM, kg), fat mass index (FMI, kg/m2), fat-free mass (FFM, kg), fat-free mass index (FFMI, kg/m2) - measured by air displacement plethysmography - and body mass index for age (BMI/age, Z-score). The exposures of interest were the gestational index (GA) of infants born at less than 33 weeks, from 34 to 36 and from 37 to 41, and intrauterine growth categorized as small (SGA), adequate (AGA) and large (LGA) for gestational age. Analysis of variance was used to compare means and linear regression was used to assess the strength of association. The analyses were adjusted according to variables collected at birth, such as monthly family income, maternal characteristics - education, age, pre-gestational body mass index (BMI), weight gain during pregnancy, smoking during pregnancy, type of delivery, and parity - and adolescent characteristics - skin color and birth weight. For analysis, FM and FMI underwent logarithmic transformation due to data asymmetry. RESULTS A total of 3,401 adolescents were analyzed, including boys and girls born at less than 33 weeks, with lower FM and FFM means than those born at term. However, in the adjusted analyses, there was no association between GA and any of the outcomes in either sex. LGA boys had a 10.5% higher FMI (p = 0.026) and +0.3 BMI/age Z-score (p = 0.019) as compared to AGA boys, and LGA girls had +0.3 kg/m 2 of FFMI (p = 0.039) than AGA girls. CONCLUSION GA was not associated with body composition at 11 years of age. However, LGA boys had higher BMI and BMI/age Z-score, and LGA girls had higher FFMI than AGA girls.
Collapse
Affiliation(s)
- Caroline Cardozo Bortolotto
- Universidade Federal de PelotasPrograma de Pós-Graduação em EpidemiologiaDepartamento de Medicina SocialPelotasRSBrasilUniversidade Federal de Pelotas. Programa de Pós-Graduação em Epidemiologia. Departamento de Medicina Social. Pelotas, RS, Brasil
| | - Iná da Silva dos Santos
- Universidade Federal de PelotasPrograma de Pós-Graduação em EpidemiologiaDepartamento de Medicina SocialPelotasRSBrasilUniversidade Federal de Pelotas. Programa de Pós-Graduação em Epidemiologia. Departamento de Medicina Social. Pelotas, RS, Brasil
| | - Juliana dos Santos Vaz
- Universidade Federal de PelotasPrograma de Pós-Graduação em EpidemiologiaDepartamento de Medicina SocialPelotasRSBrasilUniversidade Federal de Pelotas. Programa de Pós-Graduação em Epidemiologia. Departamento de Medicina Social. Pelotas, RS, Brasil
- Universidade Federal de PelotasFaculdade de NutriçãoDepartamento de NutriçãoPelotasRSBrasilUniversidade Federal de Pelotas. Faculdade de Nutrição. Departamento de Nutrição. Pelotas, RS, Brasil
| | - Alicia Matijasevich
- Universidade Federal de PelotasPrograma de Pós-Graduação em EpidemiologiaDepartamento de Medicina SocialPelotasRSBrasilUniversidade Federal de Pelotas. Programa de Pós-Graduação em Epidemiologia. Departamento de Medicina Social. Pelotas, RS, Brasil
- Universidade de São PauloFaculdade de MedicinaDepartamento de Medicina PreventivaPelotasRSBrasilUniversidade de São Paulo. Faculdade de Medicina. Departamento de Medicina Preventiva. Pelotas, RS, Brasil
| |
Collapse
|
47
|
Galea LA, Lee BH, de leon RG, Rajah MN, Einstein G. Beyond sex and gender differences: The case for women's health research. PRINCIPLES OF GENDER-SPECIFIC MEDICINE 2023:699-711. [DOI: 10.1016/b978-0-323-88534-8.00045-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Tang P, He W, Shao Y, Liu B, Huang H, Liang J, Liao Q, Tang Y, Mo M, Zhou Y, Li H, Huang D, Liu S, Zeng X, Qiu X. Associations between prenatal multiple plasma metal exposure and newborn telomere length: Effect modification by maternal age and infant sex. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120451. [PMID: 36270567 DOI: 10.1016/j.envpol.2022.120451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/14/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
Exposure to metals during pregnancy may affect maternal and infant health. However, studies on the combined effects of metals on the telomere length (TL) of newborns are limited. A prospective cohort study was conducted among 1313 mother-newborn pairs in the Guangxi Zhuang Birth Cohort. The concentrations of metals in maternal plasma during the first trimester were measured using inductively coupled plasma-mass spectrometry. We explored the associations between nine plasma metals and newborn TL using generalized linear models (GLMs), principal component analysis (PCA), quantile g-computation (qgcomp), and Bayesian kernel machine regression (BKMR). The GLMs revealed the inverse association between plasma arsenic (percent change, -5.56%; 95% CI: -7.69%, -3.38%) and barium concentrations (-9.84%; 95% CI: -13.81%, -5.68%) and newborn TL. Lead levels were related to significant decreases in newborn TL only in females. The PCA revealed a negative association between the PC3 and newborn TL (-4.52%; 95% CI: -6.34%, -2.68%). In the BKMR, the joint effect of metals was negatively associated with newborn TL. Qgcomp indicated that each one-tertile increase in metal mixture levels was associated with shorter newborn TL (-9.39%; 95% CI: -14.32%, -4.18%). The single and joint effects of multiple metals were more pronounced among pregnant women carrying female fetuses and among pregnant women <28 years of age. The finding suggests that prenatal exposure to arsenic, barium, antimony, and lead and mixed metals may shorten newborn TLs. The relationship between metal exposures and newborn TL may exhibit heterogeneities according to infant sex and maternal age.
Collapse
Affiliation(s)
- Peng Tang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wanting He
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yantao Shao
- The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530031, Guangxi, China
| | - Bihu Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huishen Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jun Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qian Liao
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ying Tang
- Department of Sanitary Chemistry, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Meile Mo
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yong Zhou
- School of Public Health, Xiangnan University, Chenzhou, 423000, China
| | - Han Li
- Department of Sanitary Chemistry, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Dongping Huang
- Department of Sanitary Chemistry, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shun Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoyun Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoqiang Qiu
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
49
|
Wolfova K, Wu D, Weiss J, Cermakova P, Kohler HP, Skirbekk VF, Stern Y, Gemmill A, Tom SE. Sons and parental cognition in mid-life and older adulthood. J Psychiatr Res 2022; 156:284-290. [PMID: 36279678 PMCID: PMC10103684 DOI: 10.1016/j.jpsychires.2022.10.026] [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: 07/05/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Prior research suggests a relationship between number of sons and maternal long-term health outcomes, including dementia. We assessed the relationship between having sons and parental cognitive aging. Specifically, we investigated the relationship between having at least 1 son and parental baseline cognition level and rate of cognitive decline, accounting for life course sociodemographic characteristics in a cohort of 13 222 adults aged ≥50 years from the US Health and Retirement Study. We included only participants with at least one child. We further explored whether this relationship varies by parental sex and whether the magnitude of the relationship increases with each additional son. Cognition was assessed biennially for a maximum of nine times as a sum of scores from immediate and delayed 10-noun free recall tests, a serial 7s subtraction test, and a backwards counting test. Associations were evaluated using linear mixed-effects models, stepwise adjusting for sociodemographic and health-related factors. In our analytic sample of parents, a total of 82.3% of respondents had at least 1 son and 61.6% of respondents were female. Parents of at least 1 son had a faster rate of cognitive decline in comparison to parents without any son. Our results also suggest that cognitive decline was faster among parents of multiple sons, compared to parents with only daughters. Thus, the results support the theory that having sons might have a long-term negative effect on parental cognition.
Collapse
Affiliation(s)
- Katrin Wolfova
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, 100 00, Czech Republic; National Institute of Mental Health, Klecany, 250 67, Czech Republic; Department of Neurology, Columbia University, 10032, New York, USA
| | - Di Wu
- Department of Biostatistics, Columbia University, 10032, New York, USA
| | - Jordan Weiss
- Department of Demography, UC Berkeley, Berkeley, CA, 94720, USA
| | - Pavla Cermakova
- National Institute of Mental Health, Klecany, 250 67, Czech Republic; Department of Epidemiology, Second Faculty of Medicine, Charles University, Prague, 150 06, Czech Republic
| | - Hans-Peter Kohler
- Department of Sociology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vegard Fykse Skirbekk
- Centre for Fertility and Health, The Norwegian Institute of Public Health, Oslo, 0473, Norway
| | - Yaakov Stern
- Department of Neurology, Columbia University, 10032, New York, USA
| | - Alison Gemmill
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, 21205, Maryland, USA
| | - Sarah E Tom
- Department of Neurology, Columbia University, 10032, New York, USA; Department of Epidemiology, Columbia University, 10032, New York, USA.
| |
Collapse
|
50
|
Freedman AN, Eaves LA, Rager JE, Gavino-Lopez N, Smeester L, Bangma J, Santos HP, Joseph RM, Kuban KC, O'Shea TM, Fry RC. The placenta epigenome-brain axis: placental epigenomic and transcriptomic responses that preprogram cognitive impairment. Epigenomics 2022; 14:897-911. [PMID: 36073148 PMCID: PMC9475498 DOI: 10.2217/epi-2022-0061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The placenta-brain axis reflects a developmental linkage where disrupted placental function is associated with impaired neurodevelopment later in life. Placental gene expression and the expression of epigenetic modifiers such as miRNAs may be tied to these impairments and are understudied. Materials & methods: The expression levels of mRNAs (n = 37,268) and their targeting miRNAs (n = 2083) were assessed within placentas collected from the ELGAN study cohort (n = 386). The ELGAN adolescents were assessed for neurocognitive function at age 10 and the association with placental mRNA/miRNAs was determined. Results: Placental mRNAs related to inflammatory and apoptotic processes are under miRNA control and associated with cognitive impairment at age 10. Conclusion: Findings highlight key placenta epigenome-brain relationships that support the developmental origins of health and disease hypothesis.
Collapse
Affiliation(s)
- Anastasia N Freedman
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lauren A Eaves
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Julia E Rager
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Noemi Gavino-Lopez
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lisa Smeester
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Bangma
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hudson P Santos
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA.,School of Nursing and Health Studies, University of Miami, Coral Gables, FL 33124, USA
| | - Robert M Joseph
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Karl Ck Kuban
- Department of Pediatrics, Division of Child Neurology, Boston Medical Center, Boston, MA 02118, USA
| | - Thomas Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rebecca C Fry
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|