1
|
Kubler JM, Beetham KS, Steane SE, Holland OJ, Borg DJ, Rae KM, Kumar S, Clifton VL. Sex-specific associations between feto-placental growth and maternal physical activity volume and sitting time: Findings from the Queensland Family Cohort study. Placenta 2024; 160:107-117. [PMID: 39787953 DOI: 10.1016/j.placenta.2024.12.027] [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: 10/10/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Antenatal physical activity (PA) is associated with beneficial changes in placental growth and function; however, the effect of excessive sitting time is less clear. The aim of this study was to investigate whether feto-placental growth changes with maternal activity, and whether these associations differ in a sex-specific manner. METHODS This study included women enrolled in the Queensland Family Cohort study who self-reported PA and sitting time at 24 or 36 weeks of gestation. Placental growth factors and feto-placental growth parameters at delivery were analysed by PA volume and sitting time, as well as by fetal sex. RESULTS Women who reported excessive sitting time during mid-pregnancy and had a female fetus showed higher placental PlGF (p = 0.031) and FLT1 (p = 0.032) mRNA expression with no difference in placental size at delivery. For the male, excessive sitting time during mid-pregnancy was associated with a lower placental weight (p = 0.001) and placental surface area (p = 0.012) and a higher birthweight to placental weight (BWPW) ratio (p = 0.042), with no change in placental growth factors. Moderate volume PA during mid-pregnancy was associated with lower VEGFA mRNA expression in the male placenta (p = 0.005) and a higher abdominal circumference in the female neonate (p = 0.042), with no overall difference in placental weight or birthweight for either sex. CONCLUSIONS The results of this study suggest that mid-pregnancy may be an important timepoint for programming of feto-placental growth in relation to maternal activity. Our findings highlight the independent benefits of reducing sitting time during pregnancy, particularly for women carrying male fetuses.
Collapse
Affiliation(s)
- Jade M Kubler
- Faculty of Medicine, Mater Research Institute-University of Queensland, South Brisbane, Australia
| | - Kassia S Beetham
- School of Behavioural and Health Sciences, Australian Catholic University, Banyo, Australia
| | - Sarah E Steane
- School of Biomedical Sciences, University of Queensland, St Lucia, Australia
| | - Olivia J Holland
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Australia
| | - Danielle J Borg
- Faculty of Medicine, Mater Research Institute-University of Queensland, South Brisbane, Australia
| | - Kym M Rae
- Faculty of Medicine, Mater Research Institute-University of Queensland, South Brisbane, Australia; Indigenous Health Research Group, Mater Research Institute, Aubigny Place, South Brisbane, Australia
| | - Sailesh Kumar
- Faculty of Medicine, Mater Research Institute-University of Queensland, South Brisbane, Australia
| | - Vicki L Clifton
- Faculty of Medicine, Mater Research Institute-University of Queensland, South Brisbane, Australia.
| |
Collapse
|
2
|
Kelly A, Chan J, Powell TL, Cox LA, Jansson T, Rosario FJ. Maternal obesity alters the placental transcriptome in a fetal sex-dependent manner. Front Cell Dev Biol 2023; 11:1178533. [PMID: 37397247 PMCID: PMC10309565 DOI: 10.3389/fcell.2023.1178533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Infants born to obese mothers have an increased risk of developing obesity and metabolic diseases in childhood and adulthood. Although the molecular mechanisms linking maternal obesity during pregnancy to the development of metabolic diseases in offspring are poorly understood, evidence suggests that changes in the placental function may play a role. Using a mouse model of diet-induced obesity with fetal overgrowth, we performed RNA-seq analysis at embryonic day 18.5 to identify genes differentially expressed in the placentas of obese and normal-weight dams (controls). In male placentas, 511 genes were upregulated and 791 genes were downregulated in response to maternal obesity. In female placentas, 722 genes were downregulated and 474 genes were upregulated in response to maternal obesity. The top canonical pathway downregulated in maternal obesity in male placentas was oxidative phosphorylation. In contrast, sirtuin signaling, NF-kB signaling, phosphatidylinositol, and fatty acid degradation were upregulated. In female placentas, the top canonical pathways downregulated in maternal obesity were triacylglycerol biosynthesis, glycerophospholipid metabolism, and endocytosis. In contrast, bone morphogenetic protein, TNF, and MAPK signaling were upregulated in the female placentas of the obese group. In agreement with RNA-seq data, the expression of proteins associated with oxidative phosphorylation was downregulated in male but not female placentas of obese mice. Similarly, sex-specific changes in the protein expression of mitochondrial complexes were found in placentas collected from obese women delivering large-for-gestational-age (LGA) babies. In conclusion, maternal obesity with fetal overgrowth differentially regulates the placental transcriptome in male and female placentas, including genes involved in oxidative phosphorylation.
Collapse
Affiliation(s)
- Amy Kelly
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeannie Chan
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Laura A. Cox
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
3
|
Merhej T, Zein JG. Epidemiology of Asthma: Prevalence and Burden of Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:3-23. [PMID: 37464114 DOI: 10.1007/978-3-031-32259-4_1] [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: 07/20/2023]
Abstract
Asthma, a common airway disease, results in a significant burden to both patients and society worldwide. Yet, despite global political commitment backed by the United Nations, progress to reduce the burden of asthma remains inadequate. This is particularly true in low-income countries. To date, progress has been delayed by the lack of uniform data collection, imperfect surveillance methods, inadequate resources, poor access to effective therapies, substandard asthma education, ineffective governmental policies, rapid urbanization, progressive increase in asthma prevalence, increased life expectancy and obesity rates worldwide, asthma heterogeneity and disease complexity, smoking, and environmental exposures to allergens and pollution. A thorough understanding of the challenges facing the international community is essential to define future strategies to improve the burden of asthma.
Collapse
Affiliation(s)
| | - Joe G Zein
- Respiratory Institute. Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
4
|
Farooq S, Khatri S. Life Course of Asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:43-76. [PMID: 37464116 DOI: 10.1007/978-3-031-32259-4_3] [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: 07/20/2023]
Abstract
Asthma is a heterogeneous chronic airway disease that can vary over a lifetime. Although broad categories of asthma by severity and type have been constructed, there remains a tremendous opportunity to discover an approach to managing asthma with additional factors in mind. Many in the field have suggested and are pursuing a novel paradigm shift in how asthma might be better managed, considering the life course of exposures, management priorities, and predicted trajectory of lung function growth. This approach will require a more holistic view of prenatal, postnatal, adolescence, hormonal and gender aspects, and the aging process. In addition, the environment, externally and internally, including in one's genetic code and epigenetic changes, are factors that affect how asthma progresses or becomes more stable in individuals. This chapter focuses on the various influences that may, to differing degrees, affect people with asthma, which can develop at any time in their lives. Shifting the paradigm of thought and strategies for care and advocating for public policies and health delivery that focus on this philosophy is paramount to advance asthma care for all.
Collapse
Affiliation(s)
- Sobia Farooq
- National Heart, Lung, and Blood Institute, CMO Division of Lung Diseases, Bethesda, MD, USA
| | - Sumita Khatri
- National Heart, Lung, and Blood Institute, CMO Division of Lung Diseases, Bethesda, MD, USA.
| |
Collapse
|
5
|
Braun AE, Mitchel OR, Gonzalez TL, Sun T, Flowers AE, Pisarska MD, Winn VD. Sex at the interface: the origin and impact of sex differences in the developing human placenta. Biol Sex Differ 2022; 13:50. [PMID: 36114567 PMCID: PMC9482177 DOI: 10.1186/s13293-022-00459-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/02/2022] [Indexed: 11/20/2022] Open
Abstract
The fetal placenta is a source of hormones and immune factors that play a vital role in maintaining pregnancy and facilitating fetal growth. Cells in this extraembryonic compartment match the chromosomal sex of the embryo itself. Sex differences have been observed in common gestational pathologies, highlighting the importance of maternal immune tolerance to the fetal compartment. Over the past decade, several studies examining placentas from term pregnancies have revealed widespread sex differences in hormone signaling, immune signaling, and metabolic functions. Given the rapid and dynamic development of the human placenta, sex differences that exist at term (37–42 weeks gestation) are unlikely to align precisely with those present at earlier stages when the fetal–maternal interface is being formed and the foundations of a healthy or diseased pregnancy are established. While fetal sex as a variable is often left unreported in studies performing transcriptomic profiling of the first-trimester human placenta, four recent studies have specifically examined fetal sex in early human placental development. In this review, we discuss the findings from these publications and consider the evidence for the genetic, hormonal, and immune mechanisms that are theorized to account for sex differences in early human placenta. We also highlight the cellular and molecular processes that are most likely to be impacted by fetal sex and the evolutionary pressures that may have given rise to these differences. With growing recognition of the fetal origins of health and disease, it is important to shed light on sex differences in early prenatal development, as these observations may unlock insight into the foundations of sex-biased pathologies that emerge later in life. Placental sex differences exist from early prenatal development, and may help explain sex differences in pregnancy outcomes. Transcriptome profiling of early to mid-gestation placenta reveals that immune signaling is a hub of early prenatal sex differences. Differentially expressed genes between male and female placenta fall into the following functional associations: chromatin modification, transcription, splicing, translation, signal transduction, metabolic regulation, cell death and autophagy regulation, ubiquitination, cell adhesion and cell–cell interaction. Placental sex differences likely reflect the interaction of cell-intrinsic chromosome complement with extrinsic endocrine signals from the fetal compartment that accompany gonadal differentiation. Understanding the mechanisms behind sex differences in placental development and function will provide key insight into molecular targets that can be modulated to improve sex-biased obstetrical complications.
Collapse
|
6
|
O'Callaghan JL, Clifton VL, Prentis P, Ewing A, Saif Z, Pelzer ES. Sex-dependent differential transcript expression in the placenta of growth restricted infants. Placenta 2022; 128:1-8. [PMID: 36031700 DOI: 10.1016/j.placenta.2022.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/03/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
INTRODUCTION The pathological decrease of fetal growth during gestation can lead to subsequent poor health outcomes for the fetus. This process is commonly controlled by the placenta, the interface between mother and baby during gestation. Sex-specific gene expression has been implicated in placental function, therefore, there is a need to determine if it is important during reduced fetal growth. We therefore aimed to characterise placental gene expression at term to evaluate sex-specific genetic changes that occur in small for gestational age (SGA) infants. METHODS RNA-sequencing of twelve human placental tissue samples collected from pregnancies yielding either term appropriate for gestational age (AGA) or SGA infants identified at delivery. Candidate genes associated with fetal size and fetal sex were identified using differential gene expression and weighted gene co-expression network analyses. Single-cell sequencing data was used for candidate validation and to estimate candidate transcript expression in specific placental cell populations. RESULTS Differential gene expression and weighted gene co-expression network analyses identified 403 candidate transcripts associated with SGA infants. One hundred and three of these transcripts showed sex-specific expression. . Published placental sequencing datasets were used to validate the key expression results from the twelve placental samples initially studied; the sex-independent transcript expression for genes involved in cell cycle processes in males (7 transcripts) and endoplasmic reticulum stress in females (17 transcripts). DISCUSSION This study identified the activation of multiple molecular mechanisms involved in the placental response to an adverse environmental stressor. Mechanisms such as disrupted protein synthesis were shared between infant biological sex when comparing AGA to SGA, whilst other pathways such as cell cycle and endoplasmic reticulum stress appear as independent/specific to either males or females when investigating reduced fetal growth. This data suggests that sexual dimorphism is an important consideration when examining placental dysfunction and poor fetal growth.
Collapse
Affiliation(s)
- Jessica L O'Callaghan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, 4001, Australia
| | - Vicki L Clifton
- Pregnancy and Development Group, Mater Research, Translational Research Institute and the University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Peter Prentis
- School of Biology and Environmental Sciences, Faculty of Science, Queensland University of Technology, Brisbane, Queensland, 4001, Australia
| | - Adam Ewing
- Pregnancy and Development Group, Mater Research, Translational Research Institute and the University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Zarqa Saif
- Pregnancy and Development Group, Mater Research, Translational Research Institute and the University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Elise S Pelzer
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, 4001, Australia.
| |
Collapse
|
7
|
Asthma in a prospective cohort of rural pregnant women from Sri Lanka: Need for better care during the pre-conceptional and antenatal period. PLoS One 2022; 17:e0269888. [PMID: 35834567 PMCID: PMC9282538 DOI: 10.1371/journal.pone.0269888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/29/2022] [Indexed: 11/29/2022] Open
Abstract
Objectives To describe the epidemiology and the effect of asthma on pregnancy outcomes in pregnant women from a rural geography. Methods We conducted a prospective cohort study in Anuradhapura district, Sri Lanka enrolling all eligible pregnant women registered in the maternal care program. An interviewer-administered questionnaire-based symptom analysis and clinical assessment was conducted in the first and second trimesters. Results We recruited 3374 pregnant women aged 15–48 years at conception. Self-reported physician-diagnosed asthma prevalence was 6.6% (n = 223) with only 41.7% (n = 93) on regular medical follow-up for asthma. The prevalence of wheeze reduced from pre-pregnancy (67.0%) to the first (46.4%) and second trimesters (47.7%; p<0.01). Of the 73 asthmatic women who did not have wheeze in the last 3 months preceding pregnancy, new-onset wheeze was reported by 6(8.2%) and 12(16.4%) in the first and second trimester, respectively. Pregnant women who sought medical care for asthma in the private sector had a lower likelihood of developing new-onset wheeze in the first trimester (p = 0.03; unadjusted OR = 0.94;95%CI 0.89–0.99). Thirty-four (33.3%) pregnant women had at least one hospital admission due to exacerbation of wheeze during the first and second trimester. The prevalence of low birth weight (16.0%) was higher among pregnant asthmatic women. Conclusion This study reports the high prevalence of asthma and asthma-associated pregnancy outcomes in women from a rural geography signifying the importance of targeted management.
Collapse
|
8
|
Lee AG, Tignor N, Cowell W, Colicino E, Bozack A, Baccarelli A, Wang P, Wright RJ. Associations between antenatal maternal asthma status and placental DNA methylation. Placenta 2022; 126:184-195. [PMID: 35858526 PMCID: PMC9679966 DOI: 10.1016/j.placenta.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Maternal asthma in pregnancy is associated with adverse perinatal and child health outcomes; however, mechanisms are poorly understood. METHODS The PRogramming of Intergenerational Stress Mechanisms (PRISM) prospective pregnancy cohort characterized asthma history during pregnancy via questionnaires and quantified placental DNAm using the Illumina Infinium HumanMethylation450 BeadChip. We performed epigenome-wide association analyses (n = 223) to estimate associations between maternal active or inactive asthma, as compared to never asthma, and placental differentially methylated positions (DMPs) and differentially variable positions (DVPs). Models adjusted for maternal pre-pregnancy body mass index, smoking status, parity, age and education level and child sex. P-values were FDR-adjusted. RESULTS One hundred and fifty-nine (71.3%) pregnant women reported no history of asthma (never asthma), 15 (6.7%) reported inactive, and 49 (22%) reported active antenatal asthma. Women predominantly self-identified as Black/Hispanic Black [88 (39.5%)] and Hispanic/non-Black [42 (18.8%)]. We identified 10 probes at FDR<0.05 and 4 probes at FDR<0.10 characterized by higher variability in maternal active asthma compared to never asthma mapping to GPX3, LHPP, PECAM1, ATAD3C, and ARHGEF4 and 2 probes characterized by lower variation mapping to CHMP4A and C5orf24. Amongst women with inactive asthma, we identified 52 probes, 41 at FDR<0.05 and an additional 11 at FDR <0.10, with higher variability compared to never asthma; BMP4, LHPP, PHYHIPL, and ZSCAN23 were associated with multiple DVPs. No associations were observed with DMPs. DISCUSSION We observed alterations in placental DNAm in women with antenatal asthma, as compared to women without a history of asthma. Further research is needed to understand the impact on fetal development.
Collapse
Affiliation(s)
- Alison G Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Nicole Tignor
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Whitney Cowell
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Bozack
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Andrea Baccarelli
- Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Andrews SV, Yang IJ, Froehlich K, Oskotsky T, Sirota M. Large-scale placenta DNA methylation integrated analysis reveals fetal sex-specific differentially methylated CpG sites and regions. Sci Rep 2022; 12:9396. [PMID: 35672357 PMCID: PMC9174475 DOI: 10.1038/s41598-022-13544-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/17/2022] [Indexed: 11/14/2022] Open
Abstract
Although male–female differences in placental structure and function have been observed, little is understood about their molecular underpinnings. Here, we present a mega-analysis of 14 publicly available placenta DNA methylation (DNAm) microarray datasets to identify individual CpGs and regions associated with fetal sex. In the discovery dataset of placentas from full term pregnancies (N = 532 samples), 5212 CpGs met genome-wide significance (p < 1E−8) and were enriched in pathways such as keratinization (FDR p-value = 7.37E−14), chemokine activity (FDR p-value = 1.56E−2), and eosinophil migration (FDR p-value = 1.83E−2). Nine differentially methylated regions were identified (fwerArea < 0.1) including a region in the promoter of ZNF300 that showed consistent differential DNAm in samples from earlier timepoints in pregnancy and appeared to be driven predominately by effects in the trophoblast cell type. We describe the largest study of fetal sex differences in placenta DNAm performed to date, revealing genes and pathways characterizing sex-specific placenta function and health outcomes later in life.
Collapse
Affiliation(s)
- Shan V Andrews
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Irene J Yang
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA.,Dougherty Valley High School, San Ramon, CA, USA
| | - Karolin Froehlich
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA. .,Department of Pediatrics, UCSF, San Francisco, CA, USA.
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA. .,Department of Pediatrics, UCSF, San Francisco, CA, USA.
| |
Collapse
|
10
|
Chatterjee S, Zeng X, Ouidir M, Tesfaye M, Zhang C, Tekola-Ayele F. Sex-specific placental gene expression signatures of small for gestational age at birth. Placenta 2022; 121:82-90. [PMID: 35303517 PMCID: PMC9010378 DOI: 10.1016/j.placenta.2022.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/14/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Small for gestational age at birth (SGA), often a consequence of placental dysfunction, is a risk factor for neonatal morbidity and later life cardiometabolic diseases. There are sex differences in placental gene expression and fetal growth. Here, we investigated sex-specific associations between gene expression in human placenta measured using RNA sequencing and SGA status using data from ethnic diverse pregnant women in the NICHD Fetal Growth Studies cohort (n = 74). METHODS Gene expression measures were obtained using RNA-Sequencing and differential gene expression between SGA (birthweight <10th percentile) and appropriate for gestational age (AGA: ≥10th and <90th percentile) was tested separately in males (12 SGA and 27 AGA) and females (9 SGA and 26 AGA) using a weighted mean of log ratios method with adjustment for mode of delivery and ethnicity. RESULTS At 5% false discovery rate (FDR), we identified 40 differentially expressed genes (DEGs) related to SGA status among males (95% up- and 5% down-regulated) and 314 DEGs among females (32.5% up- and 67.5% down-regulated). Seven female-specific DEGs overlapped with known imprinted genes (AXL, CYP24A1, GPR1, PLAGL1, CMTM1, DLX5, LY6D). The DEGs in males were significantly enriched for immune response and inflammation signaling pathways whereas the DEGs in females were enriched for organ development signaling pathways (FDR<0.05). Sex-combined analysis identified no additional DEGs, rather 98% of the sex-specific DEGs were no longer significant and the remaining 2% were attenuated. DISCUSSION This study revealed sex-specific human placental gene expression changes and molecular pathways associated with SGA and underscored that unravelling the pathogenesis of SGA warrants consideration of fetal sex as a biological variable. TRIAL REGISTRATION https://www. CLINICALTRIALS gov, Unique identifier: NCT00912132.
Collapse
Affiliation(s)
- Suvo Chatterjee
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Xuehuo Zeng
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Marion Ouidir
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Markos Tesfaye
- Section of Sensory Science and Metabolism (SenSMet), National Institute on Alcohol Abuse and Alcoholism & National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Cuilin Zhang
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, 20892, MD, USA.
| |
Collapse
|
11
|
Meakin AS, Gough M, Saif Z, Clifton VL. An ex vivo approach to understanding sex-specific differences in placental androgen signalling in the presence and absence of inflammation. Placenta 2022; 120:49-58. [DOI: 10.1016/j.placenta.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/19/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
|
12
|
Yu P, Chen Y, Ge C, Wang H. Sexual dimorphism in placental development and its contribution to health and diseases. Crit Rev Toxicol 2021; 51:555-570. [PMID: 34666604 DOI: 10.1080/10408444.2021.1977237] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
According to the Developmental Origin of Health and Disease (DOHaD), intrauterine exposure to adverse environments can affect fetus and birth outcomes and lead to long-term disease susceptibility. Evidence has shown that neonatal outcomes and the timing and severity of adult diseases are sexually dimorphic. As the link between mother and fetus, the placenta is an essential regulator of fetal development programming. It is found that the physiological development trajectory of the placenta has sexual dimorphism. Furthermore, under pathological conditions, the placental function undergoes sex-specific adaptation to ensure fetal survival. Therefore, the placenta may be an important mediator of sexual dimorphism in neonatal outcomes and adult disease susceptibility. Few systematic reviews have been conducted on sexual dimorphism in placental development and its underlying mechanisms. In this review, sex chromosomes and sex hormones, as the main reasons for sexual differentiation of the placenta, will be discussed. Besides, in the etiology of fetal-originated adult diseases, overexposure to glucocorticoids is closely related to adverse neonatal outcomes and long-term disease susceptibility. Studies have found that prenatal glucocorticoid overexposure leads to sexually dimorphic expression of placental glucocorticoid receptor isoforms, resulting in different sensitivity of the placenta to glucocorticoids, and may further affect fetal development. The present review examines what is currently known about sex differences in placental development and the underlying regulatory mechanisms of this sex bias. This review highlights the importance of placental contributions to the origins of sexual dimorphism in health and diseases. It may help develop personalized diagnosis and treatment strategies for fetal development in pathological pregnancies.
Collapse
Affiliation(s)
- Pengxia Yu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China
| | - Yawen Chen
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China
| | - Caiyun Ge
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China.,Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
13
|
Roeca C, Silva E, Barentsen C, Powell TL, Jansson T. Effects of vitrification and the superovulated environment on placental function and fetal growth in an IVF mouse model. Mol Hum Reprod 2021; 26:624-635. [PMID: 32618997 DOI: 10.1093/molehr/gaaa047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/08/2020] [Indexed: 01/15/2023] Open
Abstract
In studies of human IVF, as compared to frozen embryo transfer (ET), fresh ET is associated with smaller infants and higher risk of small for gestational age infants. Recent observations suggest that ET using vitrified embryos is associated with higher pregnancy and live birth rates compared to fresh ET, but increased rates of large for gestational age infants. The mechanisms underlying these associations are largely unknown, and available evidence suggests that the influence of IVF, vitrification and the superovulated (SO) uterine environment on placental function and fetal growth is complex. This warrants further investigation given the prevalent practice in human IVF of both fresh ET into a SO uterine environment, and vitrification with ET into a more physiologic uterine environment. Using a mouse model that closely resembles human IVF, we investigated if vitrification of IVF embryos better preserves placental function and results in better pregnancy outcomes as compared to fresh ET because of transfer into a more physiologic endometrium. We found that the SO environment, independent of vitrification status, reduced implantation rates, inhibited placental mechanistic target of rapamycin signaling and induced placental stress signaling, resulting in fetal growth restriction (1.080 ± 0.05 g estrous fresh (n = 17 litters), 1.176 ± 0.05 g estrous vitrified (n = 12), 0.771 ± 0.06 g SO fresh (n = 15), 0.895 ± 0.08 g SO vitrified (n = 10), P < 0.0001). In addition, our study suggests that vitrification impairs the developmental potential of IVF blastocysts that resulted in a significantly smaller litter size (2.6 ± 2.3 fresh estrous vs 2.5 ± 2.4 fresh SO vs 1.6 ± 1.7 estrous vitrified vs 1.7 ± 1.8 SO vitrified, P = 0.019), with no effect on fetal growth or placental function at term. Our findings suggest that vitrification may negatively impact early embryonic viability, while the SO maternal uterine environment impairs both placental development and fetal growth in IVF.
Collapse
Affiliation(s)
- C Roeca
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - E Silva
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C Barentsen
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - T L Powell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - T Jansson
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Flom JD, Chiu YHM, Cowell W, Kannan S, Ganguri HB, Coull BA, Wright RJ, Carroll K. Maternal active asthma in pregnancy influences associations between polyunsaturated fatty acid intake and child asthma. Ann Allergy Asthma Immunol 2021; 127:553-561.e3. [PMID: 34157395 DOI: 10.1016/j.anai.2021.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Studies evaluating effects of prenatal polyunsaturated fatty acid (PUFA) intake on childhood asthma reveal mixed results. Inconsistencies may result from not accounting for important modifying factors such as maternal asthma or child sex. OBJECTIVE To evaluate whether associations between prenatal PUFA intake and childhood asthma are modified by prenatal active maternal asthma or child sex in 412 mother-child dyads. METHODS Energy-adjusted prenatal dietary and supplement intakes of omega-3 (n-3) and omega-6 (n-6) PUFAs were estimated using the Block98 Food Frequency Questionnaire, administered during pregnancy. Mothers reported asthma in children followed prospectively to 4.0 plus or minus 1.7 years. Generalized additive models with smooth terms for PUFA (n-3, n-6, n-6/n-3 ratio) effects were used to investigate associations between PUFAs and child asthma, without prespecifying the form of these relationships, including effect modification by active maternal asthma or child sex. RESULTS Among mothers (40% Black, 31% Hispanic), 22% had active asthma in pregnancy; 17.5% of children developed asthma. Lower maternal n-3 PUFA intake was significantly associated with risk of childhood asthma (P = .03), in particular among children of mothers with active asthma and low n-3 PUFA intake (P = .01). This inverse association was more apparent in girls (P = .01) compared with boys (P = .30), regardless of maternal asthma status. For n-6 PUFA and the n-6/n-3 ratio, there was a lower risk of childhood asthma in the midrange of intake and increased risk at higher intake (n-6 PUFA P = .10, n-6/n-3 ratio P = .13). CONCLUSION Consideration of factors that modify effects of prenatal PUFA intake on childhood asthma has implications for designing intervention strategies tailored to impact those at greatest risk.
Collapse
Affiliation(s)
- Julie D Flom
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Yueh-Hsiu Mathilda Chiu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Whitney Cowell
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Srimathi Kannan
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Harish B Ganguri
- Department of Information Systems Security, University of Cumberlands, Williamsburg, Kentucky
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kecia Carroll
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
15
|
Meakin AS, Cuffe JSM, Darby JRT, Morrison JL, Clifton VL. Let's Talk about Placental Sex, Baby: Understanding Mechanisms That Drive Female- and Male-Specific Fetal Growth and Developmental Outcomes. Int J Mol Sci 2021; 22:6386. [PMID: 34203717 PMCID: PMC8232290 DOI: 10.3390/ijms22126386] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023] Open
Abstract
It is well understood that sex differences exist between females and males even before they are born. These sex-dependent differences may contribute to altered growth and developmental outcomes for the fetus. Based on our initial observations in the human placenta, we hypothesised that the male prioritises growth pathways in order to maximise growth through to adulthood, thereby ensuring the greatest chance of reproductive success. However, this male-specific "evolutionary advantage" likely contributes to males being less adaptable to shifts in the in-utero environment, which then places them at a greater risk for intrauterine morbidities or mortality. Comparatively, females are more adaptable to changes in the in-utero environment at the cost of growth, which may reduce their risk of poor perinatal outcomes. The mechanisms that drive these sex-specific adaptations to a change in the in-utero environment remain unclear, but an increasing body of evidence within the field of developmental biology would suggest that alterations to placental function, as well as the feto-placental hormonal milieu, is an important contributing factor. Herein, we have addressed the current knowledge regarding sex-specific intrauterine growth differences and have examined how certain pregnancy complications may alter these female- and male-specific adaptations.
Collapse
Affiliation(s)
- Ashley S. Meakin
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Vicki L. Clifton
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4000, Australia
| |
Collapse
|
16
|
Siragher E, Sferruzzi-Perri AN. Placental hypoxia: What have we learnt from small animal models? Placenta 2021; 113:29-47. [PMID: 34074553 DOI: 10.1016/j.placenta.2021.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 12/31/2022]
Abstract
Intrauterine hypoxia is a feature of pregnancy complications, both at high altitude and sea level. To understand the placental response to reduced oxygen availability, small animal models of maternal inhalation hypoxia (MIH) or reduced uterine perfusion pressure (RUPP) may be utilised. The aim of this review was to compare the findings of those studies to identify the role of oxygen availability in adapting placental structural and functional phenotypes in relation to fetal outcome. It also sought to explore the evidence for the involvement of particular genes and protein signalling pathways in the placenta in mediating hypoxia driven alterations. The data available demonstrate that both MIH and RUPP can induce placental hypoxia, which affects placental structure and vascularity, as well as glucose, amino acid, calcium and possibly lipid transport capacity. In addition, changes have been observed in HIF, VEGF, insulin/IGF2, AMPK, mTOR, PI3K and PPARγ signalling, which may be key in linking together observed phenotypes under conditions of placental hypoxia. Many different manipulations have been examined, with varied outcomes depending on the intensity, timing and duration of the insult. Some manipulations have detrimental effects on placental phenotype, viability and fetal growth, whereas in others, the placenta appears to adapt to uphold fetal growth despite the challenge of low oxygen. Together these data suggest a complex response of the placenta to reduced oxygen availability, which links to changes in fetal outcomes. However, further work is required to explore the role of fetal sex, altered maternal physiology and placental molecular mechanisms to fully understand placental responses to hypoxia and their relevance for pregnancy outcome.
Collapse
Affiliation(s)
- Emma Siragher
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
17
|
Diceglie C, Anelli GM, Martelli C, Serati A, Lo Dico A, Lisso F, Parisi F, Novielli C, Paleari R, Cetin I, Ottobrini L, Mandò C. Placental Antioxidant Defenses and Autophagy-Related Genes in Maternal Obesity and Gestational Diabetes Mellitus. Nutrients 2021; 13:nu13041303. [PMID: 33920886 PMCID: PMC8071310 DOI: 10.3390/nu13041303] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 01/07/2023] Open
Abstract
Maternal obesity and gestational diabetes mellitus (GDM) are increasing worldwide, representing risk factors for both mother and child short/long-term outcomes. Oxidative stress, lipotoxicity and altered autophagy have already been reported in obesity, but few studies have focused on obese pregnant women with GDM. Antioxidant and macro/chaperone-mediated autophagy (CMA)-related gene expressions were evaluated herein in obese and GDM placentas. A total of 47 women with singleton pregnancies delivered by elective cesarean section were enrolled: 16 normal weight (NW), 18 obese with no comorbidities (OB GDM(–)), 13 obese with GDM (OB GDM(+)). Placental gene expression was assessed by real-time PCR. Antioxidant gene expression (CAT, GPX1, GSS) decreased, the pro-autophagic ULK1 gene increased and the chaperone-mediated autophagy regulator PHLPP1 decreased in OB GDM(–) vs. NW. On the other hand, PHLPP1 expression increased in OB GDM(+) vs. OB GDM(–). When analyzing results in relation to fetal sex, we found sexual dimorphism for both antioxidant and CMA-related gene expressions. These preliminary results can pave the way for further analyses aimed at elucidating the placental autophagy role in metabolic pregnancy disorders and its potential targetability for the treatment of diabetes outcomes.
Collapse
Affiliation(s)
- Cecilia Diceglie
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
| | - Gaia Maria Anelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
| | - Anais Serati
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
| | - Fabrizia Lisso
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
| | - Francesca Parisi
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospital, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milano, Italy;
| | - Chiara Novielli
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
| | - Renata Paleari
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospital, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milano, Italy;
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy; (C.D.); (C.M.); (A.S.); (A.L.D.); (R.P.)
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy
- Correspondence: (L.O.); (C.M.); Tel.: +39-02-503-30346 (L.O.); +39-02-503-19883 (C.M.)
| | - Chiara Mandò
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università degli Studi di Milano, 20157 Milano, Italy; (G.M.A.); (F.L.); (C.N.); (I.C.)
- Correspondence: (L.O.); (C.M.); Tel.: +39-02-503-30346 (L.O.); +39-02-503-19883 (C.M.)
| |
Collapse
|
18
|
Scott H, Phillips TJ, Sze Y, Alfieri A, Rogers MF, Volpato V, Case CP, Brunton PJ. Maternal antioxidant treatment prevents the adverse effects of prenatal stress on the offspring's brain and behavior. Neurobiol Stress 2020; 13:100281. [PMID: 33344732 PMCID: PMC7739187 DOI: 10.1016/j.ynstr.2020.100281] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Maternal exposure to stress during pregnancy is associated with an increased risk of psychiatric disorders in the offspring in later life. The mechanisms through which the effects of maternal stress are transmitted to the fetus are unclear, however the placenta, as the interface between mother and fetus, is likely to play a key role. Using a rat model, we investigated a role for placental oxidative stress in conveying the effects of maternal social stress to the fetus and the potential for treatment using a nanoparticle-bound antioxidant to prevent adverse outcomes in the offspring. Maternal psychosocial stress increased circulating corticosterone in the mother, but not in the fetuses. Maternal stress also induced oxidative stress in the placenta, but not in the fetal brain. Blocking oxidative stress using an antioxidant prevented the prenatal stress-induced anxiety phenotype in the male offspring, and prevented sex-specific neurobiological changes, specifically a reduction in dendrite lengths in the hippocampus, as well as reductions in the number of parvalbumin-positive neurons and GABA receptor subunits in the hippocampus and basolateral amygdala of the male offspring. Importantly, many of these effects were mimicked in neuronal cultures by application of placental-conditioned medium or fetal plasma from stressed pregnancies, indicating molecules released from the placenta may mediate the effects of prenatal stress on the fetal brain. Indeed, both placenta-conditioned medium and fetal plasma contained differentially abundant microRNAs following maternal stress, and their predicted targets were enriched for genes relevant to nervous system development and psychiatric disorders. The results highlight placental oxidative stress as a key mediator in transmitting the maternal social stress effects on the offspring's brain and behavior, and offer a potential intervention to prevent stress-induced fetal programming of affective disorders. Social stress in pregnancy induces oxidative stress but is prevented by antioxidant. Prenatal stress induces behavioural, neuroanatomical and neurochemical changes. Maternal antioxidant treatment prevents stress-induced effects in the offspring. Maternal stress alters the balance of microRNAs secreted from the placenta. Placental oxidative stress mediates maternal social stress effects on the offspring.
Collapse
Affiliation(s)
- H Scott
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - T J Phillips
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Y Sze
- Division of Neurobiology, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - A Alfieri
- Division of Neurobiology, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - M F Rogers
- Intelligent Systems Laboratory, University of Bristol, Merchant Venturers Building, Woodland Road, Bristol, BS8 1UB, UK
| | - V Volpato
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - C P Case
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - P J Brunton
- Division of Neurobiology, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang, 314400, PR China
| |
Collapse
|
19
|
Rosario FJ, Powell TL, Gupta MB, Cox L, Jansson T. mTORC1 Transcriptional Regulation of Ribosome Subunits, Protein Synthesis, and Molecular Transport in Primary Human Trophoblast Cells. Front Cell Dev Biol 2020; 8:583801. [PMID: 33324640 PMCID: PMC7726231 DOI: 10.3389/fcell.2020.583801] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Mechanistic Target of Rapamycin Complex 1 (mTORC1) serves as positive regulator of placental nutrient transport and mitochondrial respiration. The role of mTORC1 signaling in modulating other placental functions is largely unexplored. We used gene array following silencing of raptor to identify genes regulated by mTORC1 in primary human trophoblast (PHT) cells. Seven hundred and thirty-nine genes were differentially expressed; 487 genes were down-regulated and 252 up-regulated. Bioinformatic analyses demonstrated that inhibition of mTORC1 resulted in decreased expression of genes encoding ribosomal proteins in the 60S and 40S ribosome subunits. Furthermore, down-regulated genes were functionally enriched in genes involved in eIF2, sirtuin and mTOR signaling, mitochondrial function, and glutamine and zinc transport. Stress response genes were enriched among up-regulated genes following mTORC1 inhibition. The protein expression of ribosomal proteins RPL26 (RPL26) and Ribosomal Protein S10 (RPS10) was decreased and positively correlated to mTORC1 signaling and System A amino acid transport in human placentas collected from pregnancies complicated by intrauterine growth restriction (IUGR). In conclusion, mTORC1 signaling regulates the expression of trophoblast genes involved in ribosome and protein synthesis, mitochondrial function, lipid metabolism, nutrient transport, and angiogenesis, representing novel links between mTOR signaling and multiple placental functions critical for normal fetal growth and development.
Collapse
Affiliation(s)
- Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Madhulika B. Gupta
- Department of Biochemistry, University of Western Ontario, London, ON, Canada
| | - Laura Cox
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
20
|
Examining Sex Differences in the Human Placental Transcriptome During the First Fetal Androgen Peak. Reprod Sci 2020; 28:801-818. [PMID: 33150487 DOI: 10.1007/s43032-020-00355-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/11/2020] [Indexed: 01/10/2023]
Abstract
Sex differences in human placenta exist from early pregnancy to term, however, it is unclear whether these differences are driven solely by sex chromosome complement or are subject to differential sex hormonal regulation. Here, we survey the human chorionic villus (CV) transcriptome for sex-linked signatures from 11 to 16 gestational weeks, corresponding to the first window of increasing testis-derived androgen production in male fetuses. Illumina HiSeq RNA sequencing was performed on Lexogen Quantseq 3' libraries derived from CV biopsies (n = 11 females, n = 12 males). Differential expression (DE) was performed to identify sex-linked transcriptional signatures, followed by chromosome mapping, pathway analysis, predicted protein interaction, and post-hoc linear regressions to identify transcripts that trend over time. We observe 322 transcripts DE between male and female CV from 11 to 16 weeks, with 22 transcripts logFC > 1. Contrary to our predictions, the difference between male and female expression of DE autosomal genes was more pronounced at the earlier gestational ages. In females, we found selective upregulation of extracellular matrix components, along with a number of X-linked genes. In males, DE transcripts centered on chromosome 19, with mitochondrial, immune, and pregnancy maintenance-related transcripts upregulated. Among the highest differentially expressed autosomal genes were CCRL2, LGALS13, and LGALS14, which are known to regulate immune cell interactions. Our results provide insight into sex-linked gene expression in late first and early second trimester developing human placenta and lay the groundwork to understand the mechanistic origins of sex differences in prenatal development.
Collapse
|
21
|
Meakin AS, Saif Z, Seedat N, Clifton VL. The impact of maternal asthma during pregnancy on fetal growth and development: a review. Expert Rev Respir Med 2020; 14:1207-1216. [PMID: 32825809 DOI: 10.1080/17476348.2020.1814148] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Asthma is a highly prevalent co-morbidity during pregnancy that can worsen as gestation progresses and is associated with several adverse perinatal outcomes. These adverse outcomes often result from uncontrolled asthma during pregnancy and acute asthma exacerbations that are associated with alterations in placental function and fetal growth. AREAS COVERED This paper will discuss how maternal asthma in pregnancy affects fetal growth and development which may alter future offspring health. Changes in placental function occur in a sex-specific manner in pregnancies complicated by asthma and result in differences in fetal growth and development which may influence child health. The follow up of children from mothers with asthma suggests they are at greater risk of developing asthma, have alterations in microvascular structure that may contribute to a future risk of cardiovascular disease and epigenetic modifications in immune cell function. The current evidence suggests that appropriately managed asthma during pregnancy results in normal fetal growth and development. EXPERT OPINION Clinical management of asthma during pregnancy needs significant improvement to prevent adverse outcomes for the fetus. The key to improving maternal and fetal outcomes is through education of health professionals and parents about controlling asthma during pregnancy.
Collapse
Affiliation(s)
- Ashley S Meakin
- Mater Research Institute, The University of Queensland , Brisbane, Australia
| | - Zarqa Saif
- Mater Research Institute, The University of Queensland , Brisbane, Australia
| | - Nabila Seedat
- Mater Research Institute, The University of Queensland , Brisbane, Australia
| | - Vicki L Clifton
- Mater Research Institute, The University of Queensland , Brisbane, Australia
| |
Collapse
|
22
|
An interaction between fetal sex and placental weight and efficiency predicts intrauterine growth in response to maternal protein insufficiency and gestational exposure window in a mouse model of FASD. Biol Sex Differ 2020; 11:40. [PMID: 32690098 PMCID: PMC7372829 DOI: 10.1186/s13293-020-00320-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/13/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Individuals exposed to gestational stressors such as alcohol exhibit a spectrum of growth patterns, suggesting individualized responses to the stressors. We hypothesized that intrauterine growth responses to gestational alcohol are modified not only by the stressor's severity but by fetal sex and the placenta's adaptive capacity. METHODS Pregnant C57BL/6J mice were assigned to one of three groups. Group 1 consumed a normal protein diet (18% protein by weight) and received 4.5 g alcohol/kg body weight (NP-Alc-8) or isocaloric maltodextrin (NP-MD-8) daily from embryonic day (E) 8.5-E17.5. Group 2 consumed the same diet but received alcohol (NP-Alc-13) or maltodextrin (NP-MD-13) daily from E13.5-E17.5. Group 3 consumed the same diet but containing a lower protein content (12% protein by weight) from E0.5 and also received alcohol (LP-Alc-8) or maltodextrin (LP-MD-8) daily from E8.5-E17.5. Maternal, placental, and fetal outcomes were assessed on E17.5 using 2-way ANOVA or mixed linear model. RESULTS We found that intrauterine growth differed in the alcohol-exposed fetuses depending on sex and insult severity. Both NP-Alc-8 (vs. NP-MD-8) males and females had lower body weight and asymmetrical growth, but only NP-Alc-8 females had lower placental weight (P < 0.05). NP-Alc-13 (vs. NP-MD-13) females, but not their male littermates, had lower body weight (P = 0.019). Alcohol exposure beginning from E8.5 (vs. E13.5) decreased the ratio of fetal liver-to-body weight and increased the ratio of fetal brain-to-liver weight in both sexes (P < 0.05). LP-Alc-8 (vs. NP-MD-8) group had smaller litter size (P = 0.048), but the survivors had normal placental and body weight at E17.5. Nevertheless, LP-Alc-8 fetuses still showed asymmetrical growth. Correlation analyses reveal a relationship between litter size and placental outcomes, which were related to fetal outcomes in a sex-dependent manner, suggesting that the placenta may mediate the consequence of LP-Alc-altered litter size on fetal development. CONCLUSIONS Our data indicate that the placenta is strongly involved in the fetal stress response and adapts in a sex-dependent fashion to support fetal development under the alcohol stressor. These variables may further influence the spectrum of intrauterine growth outcomes observed in those diagnosed with fetal alcohol spectrum disorder.
Collapse
|
23
|
Cissé YM, Chan JC, Nugent BM, Banducci C, Bale TL. Brain and placental transcriptional responses as a readout of maternal and paternal preconception stress are fetal sex specific. Placenta 2020; 100:164-170. [PMID: 32980048 DOI: 10.1016/j.placenta.2020.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Despite a wealth of epidemiological evidence that cumulative parental lifetime stress experiences prior to conception are determinant of offspring developmental trajectories, there is a lack of insight on how these previous stress experiences are stored and communicated intergenerationally. Preconception experiences may impact offspring development through alterations in transcriptional regulation of the placenta, a major determinant of offspring growth and sex-specific developmental outcomes. We evaluated the lasting influence of maternal and paternal preconception stress (PCS) on the mid-gestation placenta and fetal brain, utilizing their transcriptomes as proximate readouts of intergenerational impact. METHODS To assess the combined vs. dominant influence of maternal and paternal preconception environment on sex-specific fetal development, we compared transcriptional outcomes using a breeding scheme of one stressed parent, both stressed parents, or no stressed parents as controls. RESULTS Interestingly, offspring sex affected the directionality of transcriptional changes in response to PCS, where male tissues showed a predominant downregulation, and female tissues showed an upregulation. There was also an intriguing effect of parental sex on placental programming where paternal PCS drove more effects in female placentas, while maternal PCS produced more transcriptional changes in male placentas. However, in the fetal brain, maternal PCS produced overall more changes in gene expression than paternal PCS, supporting the idea that the intrauterine environment may have a larger overall influence on the developing brain than it does on shaping the placenta. DISCUSSION Preconception experiences drive changes in the placental and the fetal brain transcriptome at a critical developmental timepoint. While not determinant, these altered transcriptional states may underlie sex-biased risk or resilience to stressful experiences later in life.
Collapse
Affiliation(s)
- Yasmine M Cissé
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Jennifer C Chan
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Bridget M Nugent
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Caitlin Banducci
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Tracy L Bale
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States.
| |
Collapse
|
24
|
Saoi M, Kennedy KM, Gohir W, Sloboda DM, Britz-McKibbin P. Placental Metabolomics for Assessment of Sex-specific Differences in Fetal Development During Normal Gestation. Sci Rep 2020; 10:9399. [PMID: 32523064 PMCID: PMC7286906 DOI: 10.1038/s41598-020-66222-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
The placenta is a metabolically active interfacial organ that plays crucial roles in fetal nutrient delivery, gas exchange and waste removal reflecting dynamic maternal and fetal interactions during gestation. There is growing evidence that the sex of the placenta influences fetal responses to external stimuli in utero, such as changes in maternal nutrition and exposure to environmental stressors. However, the exact biochemical mechanisms associated with sex-specific metabolic adaptations during pregnancy and its link to placental function and fetal development remain poorly understood. Herein, multisegment injection-capillary electrophoresis-mass spectrometry is used as a high throughput metabolomics platform to characterize lyophilized placental tissue (~2 mg dried weight) from C57BL/6J mice fed a standardized diet. Over 130 authentic metabolites were consistently measured from placental extracts when using a nontargeted metabolomics workflow with stringent quality control and robust batch correction. Our work revealed distinct metabolic phenotype differences that exist between male (n = 14) and female (n = 14) placentae collected at embryonic day E18.5. Intracellular metabolites associated with fatty acid oxidation and purine degradation were found to be elevated in females as compared to male placentae (p < 0.05, effect size >0.40), including uric acid, valerylcarnitine, hexanoylcarnitine, and 3-hydroxyhexanolycarnitine. This murine model sheds new insights into sex-specific differences in placental mitochondrial function and protective mechanisms against deleterious oxidative stress that may impact fetal growth and birth outcomes later in life.
Collapse
Affiliation(s)
- Michelle Saoi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Wajiha Gohir
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.,Department of Pediatrics and Obstetrics and Gynecology, McMaster University, Hamilton, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada.
| |
Collapse
|
25
|
Huq F, Obida M, Bornman R, Di Lenardo T, Chevrier J. Associations between prenatal exposure to DDT and DDE and allergy symptoms and diagnoses in the Venda Health Examination of Mothers, Babies and their Environment (VHEMBE), South Africa. ENVIRONMENTAL RESEARCH 2020; 185:109366. [PMID: 32299029 PMCID: PMC7336873 DOI: 10.1016/j.envres.2020.109366] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 06/11/2023]
Abstract
Dichlorodiphenyl trichloroethane (DDT) is an organochlorine insecticide that is banned internationally except for use as part of Indoor Residual Spraying (IRS) programs to control malaria. Although animal studies show that DDT and its breakdown product dichlorodiphenyl dichloroethylene (DDE) affect the immune system and may cause allergies, no studies have examined this question in populations where IRS is conducted. The aim of our study was to investigate whether prenatal exposure to DDT and DDE is associated with allergy symptoms and diagnose among South African children living in an area where IRS is conducted. To accomplish this aim, we used data from the Venda Health Examination of Mothers, Babies and their Environment (VHEMBE), an ongoing birth cohort study of 752 children born between 2012 and 2013 in the rural Vhembe district of Limpopo, South Africa. We measured maternal peripartum serum concentrations of DDT and DDE, and administered a questionnaire to the caregivers of 658 children aged 3.5 years to collect information on allergy symptoms and diagnoses as well as potential confounders using validated instruments. Using multiple logistic regression models, we found positive associations between DDT and DDE serum concentrations and most of the allergy symptoms and diagnoses. Maternal DDT (Odds Ratio [OR] = 1.5 per 10-fold increase, 95% Confidence interval, CI = 1.0, 2.3) and DDE (OR = 1.4, 95% CI = 0.8, 2.4) serum concentrations were most strongly associated with caregiver report of wheezing or whistling in the chest. Concentrations of DDT and/or DDE were also associated with increased odds of children's chests sounding wheezy during or after exercise, itchy rashes coming and going for at least six months, diagnosis of food allergy, and diagnosis of dust or dust mites allergy but confidence intervals crossed the null. Results suggest that prenatal exposure to DDT, and possibly DDE, is associated with elevated odds of wheezing among children from an IRS area.
Collapse
Affiliation(s)
- Fahmida Huq
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Muvhulawa Obida
- University of Pretoria School of Health Systems and Public Health, and Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Riana Bornman
- University of Pretoria School of Health Systems and Public Health, and Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Thomas Di Lenardo
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
26
|
Jones AR, Tuckwell C, Wright IMR, Morrison JL, Kandasamy Y, Wittwer MR, Arstall MA, Stark MJ, Davies M, Hurst C, Okano S, Clifton VL. The impact of maternal asthma during pregnancy on offspring retinal microvascular structure and its relationship to placental growth factor production in utero. Microcirculation 2020; 27:e12622. [PMID: 32330353 DOI: 10.1111/micc.12622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022]
Abstract
Asthma is a common chronic disease in pregnancy that affects placental function and fetal growth and associated with cardio-metabolic disorders in the offspring but the mechanisms are unknown. This study explored whether maternal asthma in pregnancy is associated with the development of offspring microvascular structure and whether it was related to biomarkers of angiogenesis in utero. Children aged 4 to 6 years, born to either asthmatic mothers (n = 38) or healthy controls (n = 25), had their retinal microvascular structure examined. Maternal plasma PlGF concentrations at 18 and 36 weeks' gestation were measured. There was a significant global difference in all retinal microvascular measures between children of asthmatic mothers relative to controls and increased retinal venular tortuosity in children born to asthmatic mothers (7.1 (95% CI 0.7-13.5); P = .031). A rise in plasma PlGF from 18 to 36 weeks' gestation was observed in the control population which was significantly lower in the asthma group by 190.9 pg/mL. PlGF concentrations were correlated with microvascular structure including arteriolar branching and venular tortuosity. These exploratory findings indicate that exposure to maternal asthma during pregnancy is associated with persistent changes in microvascular structure in childhood that may be driven by alterations to angiogenic mechanisms in utero.
Collapse
Affiliation(s)
- Ailee R Jones
- Mater Medical Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Christine Tuckwell
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Ian M R Wright
- Illawarra Health and Medical Research Institute and School of Medicine, The University of Wollongong, Wollongong, NSW, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | | | - Melanie R Wittwer
- Lyell McEwin Hospital, University of Adelaide, Adelaide, SA, Australia
| | | | - Michael J Stark
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Michael Davies
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Cameron Hurst
- Statistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Satomi Okano
- Statistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Vicki L Clifton
- Mater Medical Research Institute, University of Queensland, Brisbane, QLD, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
27
|
Weinheimer C, Wang H, Comstock JM, Singh P, Wang Z, Locklear BA, Goodwin KL, Maschek JA, Cox JE, Baack ML, Joss-Moore LA. Maternal Tobacco Smoke Exposure Causes Sex-Divergent Changes in Placental Lipid Metabolism in the Rat. Reprod Sci 2020; 27:631-643. [PMID: 32046449 PMCID: PMC7539808 DOI: 10.1007/s43032-019-00065-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/01/2019] [Indexed: 12/30/2022]
Abstract
Maternal tobacco smoke exposure (MTS) affects fetal acquisition of long-chain polyunsaturated fatty acids (LCPUFA) and increases the risk of obesity and cardio-metabolic disease in the offspring. Alterations in fetal LCPUFA acquisition in maternal smoking are mediated by the placenta. The handling of LCPUFA by the placenta involves protein-mediated transfer and storage. Molecular mediators of placental LCPUFA handling include PPARγ and the fatty acid transport proteins. We previously demonstrated, in a rat model, that MTS results in programming of adult-onset obesity and metabolic disease in male, but not female, offspring. In this study, we test the hypothesis that in utero MTS exposure alters placental structure, placental LCPUFA handling, and fetal fatty acid levels, in a sex-divergent manner. We exposed pregnant rats to tobacco smoke from embryonic day 11 to term gestation. We measured placental and fetal fatty acid profiles, the systolic/diastolic ratio (SD ratio), placental histology, and expression of molecular mediators in the placenta. Our primary finding is that MTS alters fatty acid profiles in male, but not female fetuses and placenta, including increasing the ratio of omega-6 to omega-3 fatty acids. MTS also increased SD ratio in male, but not female placenta. In contrast, the expression of PPARγ and FATPs was upregulated in female, but not male placenta. We conclude that MTS causes sex-divergent changes in placental handling of LCPUFA in the rat. We speculate that our results demonstrate an adaptive response to MTS by the female placenta.
Collapse
Affiliation(s)
- Claudia Weinheimer
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Haimei Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | | | - Purneet Singh
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Zhengming Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Brent A Locklear
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Kasi L Goodwin
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - J Alan Maschek
- Health Science Center Cores, University of Utah Health Sciences Center, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - James E Cox
- Health Science Center Cores, University of Utah Health Sciences Center, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | | | - Lisa A Joss-Moore
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
28
|
Steyn A, Crowther NJ, Norris SA, Rabionet R, Estivill X, Ramsay M. Epigenetic modification of the pentose phosphate pathway and the IGF-axis in women with gestational diabetes mellitus. Epigenomics 2019; 11:1371-1385. [PMID: 31583916 DOI: 10.2217/epi-2018-0206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Gestational diabetes mellitus (GDM) has been linked with adverse long-term health outcomes for the fetus and mother. These effects may be mediated by epigenetic modifications. Materials & methods: Genome-wide RNA sequencing was performed in placental tissue and maternal blood in six GDM and six non-GDM pregnancies. Promoter region DNA methylation was examined for selected genes and correlated with gene expression to examine an epigenetic modulator mechanism. Results: Reductions of mRNA expression and increases in promoter methylation were observed for G6PD in GDM women, and for genes encoding IGF-binding proteins in GDM-exposed placenta. Conclusion: GDM involves epigenetic attenuation of G6PD, which may lead to hyperglycemia and oxidative stress, and the IGF-axis, which may modulate fetal macrosomia.
Collapse
Affiliation(s)
- Angela Steyn
- Division of Human Genetics, National Health Laboratory Service and the School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Nigel J Crowther
- Division of Human Genetics, National Health Laboratory Service and the School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa.,The Department of Chemical Pathology, School of Pathology, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Shane A Norris
- Developmental Pathways for Health Research Unit, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Raquel Rabionet
- The Centre for Genomic Regulation, Genes and Diseases Program, Barcelona, Spain
| | - Xavier Estivill
- The Centre for Genomic Regulation, Genes and Diseases Program, Barcelona, Spain
| | - Michèle Ramsay
- Division of Human Genetics, National Health Laboratory Service and the School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa.,The Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Asthma, a common respiratory disease that affects about 10% of the US population, represents a significant public health issue. In the last decade, cumulative evidence has demonstrated sex disparities in asthma, including significant differences in epidemiology, clinical presentation, response to therapies, and health outcomes. Understanding sex-related differences in asthma enables clinicians to provide personalized asthma care and improve asthma outcome. RECENT FINDINGS Recent studies on sex-related differences in asthma inform us on mechanism underlying asthma pathogenesis across all age groups. Sex hormones directly modulate immune pathways crucial in asthma pathogenesis and affect individual's response to environmental triggers and medications, such as leukokotriene inhibitors. Not surprisingly, the use of external sex hormone supplementations appears to modulate asthma risk. Identification of sex-specific asthma risk loci through genome-wide association studies also provides supporting evidence on sex-related differences in asthma. There is an interaction between sex and obesity, an interaction that could place females at higher risk for systemic inflammation and, consequently, asthma. In this article, we review epidemiological and clinical studies on sex-related differences in asthma, with a special focus on the role of sex hormones, including hormonal therapies and the asthma-obesity interaction.
Collapse
Affiliation(s)
- Peng Zhang
- From the Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Joe Zein
- From the Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
30
|
Katzman PJ, Blitman J, Metlay LA. Basal Chronic Villitis and Disorders of the Placental Basal Plate: A Possible Immunological Link Between Hypertensive Disorders of Pregnancy and Morbidly Adherent Placenta. Pediatr Dev Pathol 2019; 22:334-339. [PMID: 30665335 DOI: 10.1177/1093526619825708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDP) are a common cause for preterm delivery. Prior studies showed that chronic villitis (CV) is associated with intrauterine growth restriction, preeclampsia, intrauterine fetal death, and morbidly adherent placenta (MAP). The authors hypothesize that disorders of the placental basal plate, especially basal chronic villitis (BCV), are associated with HDP. METHODS The laboratory information system was queried over 12 years to identify placentas with or without the clinical history of HDP and with or without multifocal/focal CV or BCV. As a control for tissue sampling, a similar search was performed over 5 years for placentas evaluated for MAP. RESULTS Of 19,683 placentas identified, 14.8% had CV which was in 18.5% and 14.2% of placentas associated with or without HDP, respectively, a significant difference (P < .0001). BCV was present in 6.0% and 3.9% of placentas with or without HDP, respectively, also a significant difference (P < .0001). BCV was more likely than multifocal/focal CV to occur in HDP (32.4% vs 27.4%) when all cases of CV were analyzed (P = .025). Of 221 placentas with MAP, 64% had multifocal/focal CV and 36% had BCV. CONCLUSIONS BCV and CV are more common in placentas with HDP than in normotensive pregnancies. They are also seen in MAP, as supported by another recent study.
Collapse
Affiliation(s)
- Philip J Katzman
- 1 Division of Surgical Pathology, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Joseph Blitman
- 1 Division of Surgical Pathology, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Leon A Metlay
- 1 Division of Surgical Pathology, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
31
|
Mirzakhani H, Carey VJ, Zeiger R, Bacharier LB, O’Connor GT, Schatz MX, Laranjo N, Weiss ST, Litonjua AA. Impact of parental asthma, prenatal maternal asthma control, and vitamin D status on risk of asthma and recurrent wheeze in 3-year-old children. Clin Exp Allergy 2019; 49:419-429. [PMID: 30461089 PMCID: PMC6438751 DOI: 10.1111/cea.13320] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND While familial clustering of asthma is known, few studies have reported on the relative roles of paternal and maternal asthma and the role of maternal asthma control in pregnancy on the risk for asthma in the child. OBJECTIVE We aimed to investigate the relative roles of paternal asthma, maternal asthma, and maternal asthma control during pregnancy on the risk of asthma or recurrent wheeze in 3-year-old children and how prenatal and cord blood vitamin D status might affect this risk. METHODS Data from 806 women, their partners (biologic fathers of the infants), and their children participated in the Vitamin D Antenatal Asthma Reduction Trail (VDAART, clinicaltrials.gov identification number NCT00920621) were used for this cohort analysis. The parental report of physician-diagnosed asthma or recurrent wheeze in offspring was the main outcome. Weibull regression models for interval-censored event times were used to estimate the main variables of interests and additional covariates on the outcome. RESULTS The highest risk was observed among children with both parents being asthmatic relative to non-asthmatic parents (aHR = 2.30, 95% CI: 1.35-3.84), and less so if only the mother was asthmatic (aHR = 1.70, 95% CI: 1.17-2.40). In the subset of children born to asthmatic mothers, the risk for asthma was higher in those who were born to mothers whose asthma was uncontrolled (aHR = 1.60, 95% CI: 1.02-2.54). Children whose mothers had sufficient vitamin D status (25Hydroxyvitamin D ≥ 30 ng/mL) at early and late pregnancy and had cord blood vitamin D sufficiency demonstrated a lower risk of asthma/recurrent wheeze than children who had insufficient cord blood vitamin D status at birth (aHR = 0.47, 95% CI: 0.27-0.83). CONCLUSION AND CLINICAL RELEVANCE Careful attention to maternal asthma control, monitoring vitamin D status, and correcting insufficiency at early pregnancy and maintaining the sufficiency status throughout pregnancy have potential preventive roles in offspring asthma or recurrent wheeze.
Collapse
Affiliation(s)
- Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vincent J. Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Robert Zeiger
- Department of Allergy and Research and Evaluation, Kaiser Permanente Southern California Region, San Diego and Pasadena, California
| | - Leonard B. Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University, St. Louis, Missouri
| | - George T. O’Connor
- Pulmonary Center, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Michael X. Schatz
- Department of Allergy and Research and Evaluation, Kaiser Permanente Southern California Region, San Diego and Pasadena, California
| | - Nancy Laranjo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Partners Center for Personalized Medicine, Partners Health Care, Boston, MA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
32
|
Hicks LM, Swales DA, Garcia SE, Driver C, Davis EP. Does Prenatal Maternal Distress Contribute to Sex Differences in Child Psychopathology? Curr Psychiatry Rep 2019; 21:7. [PMID: 30729361 DOI: 10.1007/s11920-019-0992-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Prenatal maternal psychological distress is an established risk factor for the development of psychopathology in offspring. The purpose of this review is to evaluate whether sex differences in fetal responses to maternal distress contribute to sex differences in subsequent psychopathology. RECENT FINDINGS Male and female fetuses respond differently to stress signals. We review recent evidence that demonstrates a sex-specific pattern of association between prenatal maternal distress and pathways associated with risk for psychopathology including offspring hypothalamic pituitary adrenocortical (HPA) axis regulation, brain development, and negative emotionality. Prenatal maternal distress exerts sex-specific consequences on the fetus. These differences may contribute to the well-established sex differences in psychopathology and in particular to greater female vulnerability to develop internalizing problems.
Collapse
Affiliation(s)
- Laurel M Hicks
- Department of Psychology, University of Denver, 2155 South Race Street, Denver, CO, 80208, USA
| | - Danielle A Swales
- Department of Psychology, University of Denver, 2155 South Race Street, Denver, CO, 80208, USA
| | - Sarah E Garcia
- Department of Psychology, University of Denver, 2155 South Race Street, Denver, CO, 80208, USA
| | - Camille Driver
- Department of Psychology, University of Denver, 2155 South Race Street, Denver, CO, 80208, USA
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, 2155 South Race Street, Denver, CO, 80208, USA. .,Department of Psychiatry and Human Behavior, University of California Irvine, One University Drive, Orange, CA, 92866, USA.
| |
Collapse
|
33
|
Shallie PD, Naicker T. The placenta as a window to the brain: A review on the role of placental markers in prenatal programming of neurodevelopment. Int J Dev Neurosci 2019; 73:41-49. [PMID: 30634053 DOI: 10.1016/j.ijdevneu.2019.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND During development, the placenta can be said to be the most important organ, however, the most poorly researched. There is currently a broader understanding of how specific insults during development affect the fetal brain, and also the importance of placental signaling in neurodevelopmental programming. Epigenetic responses to maternal and fetal signals are an obvious candidate for transforming early life inputs into long-term programmatic outcomes. As a mediator of maternal and environmental signals to the developing fetus, epigenetic processes within the placenta are particularly powerful such that alterations of placental gene expression, downstream function, and signalling during foetal development have the potential for dramatic changes in developmental programming. SUMMARY In this article, we reviewed emerging evidence for a placental role in prenatal neurodevelopmental programming with a specific focus on nutrient and prenatal stress signals integration into chromatin changes; this new understanding, we hope will provide the means for lowering developmentally based disorder risk, and new therapeutic targets for treatment in adulthood. KEY MESSAGES Based on this review, the placenta is a potent micro-environmental player in neurodevelopment as it orchestrates a series of complex maternal-foetal interactions. Maternal insults to this microenvironment will impair these processes and disrupt foetal brain development resulting in the prenatal programming of neurodevelopmental disorders. These findings should inspire advance animal model and human research drive to appraise gene-environment impacts during pregnancy that will target the developmental cause of adult-onset mental disorders.
Collapse
Affiliation(s)
- Philemon Dauda Shallie
- Optics and Imaging Centre, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa.
| | - Thajasvarie Naicker
- Optics and Imaging Centre, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
34
|
Mirzakhani H, Carey VJ, McElrath TF, Qiu W, Hollis BW, O’Connor GT, Zeiger RS, Bacharier L, Litonjua AA, Weiss ST. Impact of Preeclampsia on the Relationship between Maternal Asthma and Offspring Asthma. An Observation from the VDAART Clinical Trial. Am J Respir Crit Care Med 2019; 199:32-42. [PMID: 30153046 PMCID: PMC6353019 DOI: 10.1164/rccm.201804-0770oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/27/2018] [Indexed: 01/27/2023] Open
Abstract
RATIONALE Maternal asthma and preeclampsia have independently been reported to be associated with increased asthma incidence in children of affected mothers. Maternal asthma is also associated with increased risk of preeclampsia development. However, the joint effect of these maternal conditions on child asthma risk is unknown. OBJECTIVES To study whether development of preeclampsia among pregnant women with asthma was associated with higher risk of childhood asthma in the VDAART (Vitamin D Antenatal Asthma Reduction Trial). METHODS A total of 806 pregnant women and their offspring at high risk of asthma or atopy, who were followed from VDAART enrollment (10-18 wk of gestation) through the child's third birthday, were included in this cohort analysis. Preeclampsia status was determined by chart review, obstetrician diagnosis, and adjudication by a panel of obstetricians. Child asthma was the main outcome as determined by parental report of a physician diagnosis, and the risk of child asthma was also examined if accompanied by recurrent wheeze. The main risk variable of interest was a four-level ordered variable defined for each mother, with values without asthma without preeclampsia, without asthma with preeclampsia, with asthma without preeclampsia, and with asthma with preeclampsia during their pregnancy. We examined the trend of outcome proportions across these categories. To account for differences in maternal and child characteristics, we used a Weibull regression model for interval-censored data to compare the incidence of child asthma by age of 3 years across the maternal variable categories. MEASUREMENTS AND MAIN RESULTS The incidence of asthma in 3-year-old children was 9.90% (44/445), 17.95% (7/39), 22.11% (65/294), and 32.14% (9/28) among those born to mothers without asthma and without preeclampsia, mothers without asthma with preeclampsia, mothers with asthma without preeclampsia, and mothers with asthma with preeclampsia, respectively. The incidences demonstrated an increasing trend in risk of child asthma across the maternal groups (P for trend <0.001). After accounting for potential confounders and using time to report of childhood asthma as analysis outcome, risk of asthma was greater among children born to mothers with asthma without preeclampsia, compared with mothers without asthma without preeclampsia (adjusted hazard ratio, 2.18; 95% confidence interval, 1.46-3.26). This risk was 50% greater for children born to mothers with asthma who developed preeclampsia during pregnancy (adjusted hazard ratio, 2.68; 95% confidence interval, 1.30-5.61). The trend in asthma and recurrent wheeze proportions across the maternal groups' children also indicated a higher risk for children born to mothers with asthma with preeclampsia (adjusted hazard ratio, 4.73; 95% confidence interval, 2.20-10.07; P for trend <0.001). CONCLUSIONS Preeclampsia is associated with increased risk of early life childhood asthma in children less than 3 years old over and above that associated with maternal asthma alone. The results implicate the interplay between maternal factors as strong predictors of offspring asthma and in utero maternal-fetal immune perturbations and developmental dysregulations associated with preeclampsia.
Collapse
Affiliation(s)
- Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vincent J. Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas F. McElrath
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Weiliang Qiu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bruce W. Hollis
- Department of Pediatrics, Medical College of South Carolina, Charleston, South Carolina
| | - George T. O’Connor
- Pulmonary Center, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Robert S. Zeiger
- Department of Allergy and Research and Evaluation, Kaiser Permanente Southern California, Region, San Diego and Pasadena, California
| | - Leonard Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University, St. Louis, Missouri
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, New York; and
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Partners Center for Personalized Medicine, Partners Health Care, Boston, Massachusetts
| |
Collapse
|
35
|
Ram S, Howland MA, Sandman CA, Davis EP, Glynn LM. Prenatal Risk for ASD: Fetal Cortisol Exposure Predicts Child Autism-Spectrum Disorder Symptoms. Clin Psychol Sci 2018; 7:349-361. [PMID: 33758678 DOI: 10.1177/2167702618811079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The etiology of autism spectrum disorder (ASD) is multifactorial and complex and likely involves interactions among genetic, epigenetic and environmental factors. With respect to environmental influences, a growing literature implicates intrauterine experiences in the origin of this pervasive developmental disorder. In this prospective longitudinal design, we examine the hypothesis that fetal exposure to maternal cortisol may confer ASD risk. In addition, because ASD is four times more prevalent in males than females and because sexually dimorphic responses to intrauterine experiences are commonly observed, we examine whether or not any associations differ by fetal sex. Maternal plasma cortisol was measured at 15, 19, 25, 31, and 37 weeks' gestation in a sample of 84 pregnant women. ASD symptoms were assessed in their 5-year old children with the Social Communication Questionnaire (SCQ). Fetal exposure to lower levels of maternal cortisol was associated with higher levels of ASD symptoms among boys only. The observed hypocortisolemic profile exhibited by these mothers may indicate a risk factor that precedes the stress of caregiving for a child with ASD and may not be solely a consequence of the stress of caregiving as previously thought. Further, these findings confirm the value of examining prenatal hormone exposures as predictors of ASD risk and support the premise that altered prenatal steroid exposures may play a role in the etiology of ASD.
Collapse
Affiliation(s)
- Sheena Ram
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, USA
| | - Mariann A Howland
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Curt A Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Elysia Poggi Davis
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA.,Department of Psychology, University of Denver, Denver, CO, USA
| | - Laura M Glynn
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA.,Department of Psychology, Chapman University, Orange, CA USA
| |
Collapse
|
36
|
Zein JG, Denson JL, Wechsler ME. Asthma over the Adult Life Course: Gender and Hormonal Influences. Clin Chest Med 2018; 40:149-161. [PMID: 30691709 DOI: 10.1016/j.ccm.2018.10.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Asthma is a common disorder that affects genders differently across the life span. Earlier in life, it is more common in boys. At puberty, asthma becomes more common and often more severe in girls and women. The effect of sex hormones on asthma incidence and its severity is difficult to differentiate from other asthma severity risk factors, such as racial background, socioeconomic factors, obesity, atopy, environmental exposure, and, in particular, lung aging. Recognizing gender-associated and age-associated differences is important to understanding the pathobiology of asthma and to providing effective education and personalized care for patients with asthma across the life course.
Collapse
Affiliation(s)
- Joe G Zein
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44106, USA
| | - Joshua L Denson
- National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | | |
Collapse
|
37
|
Risk of childhood cerebral palsy following prenatal exposure to ß2-adrenergic receptor agonist: A nationwide cohort study. PLoS One 2018; 13:e0202078. [PMID: 30114199 PMCID: PMC6095523 DOI: 10.1371/journal.pone.0202078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 07/29/2018] [Indexed: 11/19/2022] Open
Abstract
Background Cerebral palsy (CP) is the most common physical developmental disability in childhood with a prevalence of 2 to 3 per 1000 live births. β2-adrenoreceptor agonist (β2AA) are widely used for the treatment of asthma. Maternal use of β2AAs may increase the risk of adverse neuro-psychiatric health outcomes in the offspring. No study, however, has evaluated the effect of prenatal exposure to β2AAs on the risk of CP. Objective To examine the association between prenatal exposure to β2AAs and the risk of childhood cerebral palsy. Methods This population-based cohort study included all live singleton births in Denmark from January 1, 1997 to December 31, 2003. The information on outpatient prescriptions of β2AAs was extracted from Danish National Prescription Registry. Children born to mothers who used β2AAs from 30 days before pregnancy until delivery were categorized as the exposed. To differentiate the effect of β2AAs from the underlying indications, the exposure window was further extended to 2 years before pregnancy and the exposed groups were re-defined to represent different periods of exposure to maternal use of β2AAs (use only before pregnancy, use only during pregnancy, and use both before and during pregnancy). Cases of CP were identified from the Danish Cerebral Palsy Register. Logistic regression was used to estimate incidence odds ratio (OR) of CP. Results Among all the 442,278 singletons, 19,616 (4.44%) were exposed to β2AAs in utero (from 30 days before pregnancy until delivery). The risk of childhood CP was 0.21% in exposed and 0.19% in unexposed group, yielding an adjusted OR (aOR) 1.12 (95% confidence interval (CI): 0.82, 1.53). When extending the exposure time window to 2 years prior to pregnancy, no overall significant association was observed regardless of the exposure period. However, an increased risk of CP (aOR = 1.41, 95%CI: 0.92, 2.18) for maternal β2AAs use during pregnancy was observed in female offspring, especially in those born at term (aOR = 1.65, 95%CI: 1.02, 2.67). This increase was mainly attributed to an increased risk in those born to mothers who used β2AAs both before and during pregnancy (aOR = 1.81, 95%CI: 0.99, 3.33). Conclusions We observed an association between maternal β2AAs use during pregnancy and an increased risk of CP in female offspring, but we could not rule out confounding by the underlying indications for β2AAs.
Collapse
|
38
|
Braun F, Hardt AK, Ehrlich L, Sloboda DM, Challis JRG, Plagemann A, Henrich W, Braun T. Sex-specific and lasting effects of a single course of antenatal betamethasone treatment on human placental 11β-HSD2. Placenta 2018; 69:9-19. [PMID: 30213491 DOI: 10.1016/j.placenta.2018.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 01/03/2023]
Abstract
INTRODUCTION We have previously shown that even a single course of antenatal betamethasone (BET) as an inductor for lung maturity reduces birth weight and head circumference. Moreover, animal studies link BET administration to alterations of the hypothalamic-pituitary-adrenal-gland-axis (HPA). The unhindered development of the fetal HPA axis is dependent on the function and activity of 11β-hydroxysteroiddehydrogenase type 2 (11β-HSD2), a transplacental cortisol barrier. Therefore, we investigated the effects of BET on this transplacental barrier and fetal growth. METHODS Pregnant women treated with a single course of BET between 23 + 5 to 34 + 0 weeks of gestation were compared to gestational-age-matched controls. Placental size and neonatal anthropometrics were taken. Cortisol and ACTH levels were measured in maternal and umbilical cord blood samples. Placental 11β-hydroxysteroiddehydrogenase type 1 (11β-HSD1) protein levels and 11β-HSD2 protein and activity levels were determined. Parameters were analyzed independent of sex, and in subgroups divided by gender and gestational age. RESULTS In term born females, BET administration was associated with reduced head circumference and decreased 11β-HSD2 protein levels and enzyme activity. Males treated with BET, especially those born prematurely, showed increased 11β-HSD2 protein levels. CONCLUSION A single course of BET alters placental glucocorticoid metabolism in a sex-specific manner. Decreased 11β-HSD2 levels in term born females may lead to an increased placental transfer of maternal cortisol and therefore result in a reduced head circumference and a higher risk for altered stress response in adulthood. Further research is needed to conclude the significance of increased 11β-HSD2 levels in males.
Collapse
Affiliation(s)
- F Braun
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - A K Hardt
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - L Ehrlich
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - D M Sloboda
- Department of Biochemistry and Biomedical Sciences, Obstetrics & Gynecology and Pediatrics, Farncombe Family Digestive Health Research Institute, McMaster University, Main Street West, Ontario L8S4L8, Hamilton, 1280, Canada
| | - J R G Challis
- Department of Physiology and Obstetrics and Gynecology, University of Toronto King's College Circle, Ontario M5S 1A8, Toronto, Canada; Faculty of Health Sciences, Simon Fraser University, University Drive, 8888, B.C, V5A 1S6, Burnaby, Canada
| | - A Plagemann
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - W Henrich
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - T Braun
- Department of Obstetrics and Division of 'Experimental Obstetrics̔, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
39
|
Muralimanoharan S, Gao X, Weintraub S, Myatt L, Maloyan A. Sexual dimorphism in activation of placental autophagy in obese women with evidence for fetal programming from a placenta-specific mouse model. Autophagy 2018; 12:752-69. [PMID: 26986453 DOI: 10.1080/15548627.2016.1156822] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The incidence of maternal obesity and its co-morbidities (diabetes, cardiovascular disease) continues to increase at an alarming rate, with major public health implications. In utero exposure to maternal obesity has been associated with development of cardiovascular and metabolic diseases in the offspring as a result of developmental programming. The placenta regulates maternal-fetal metabolism and shows significant changes in its function with maternal obesity. Autophagy is a cell-survival process, which is responsible for the degradation of damaged organelles and misfolded proteins. Here we show an activation of autophagosomal formation and autophagosome-lysosome fusion in placentas of males but not females from overweight (OW) and obese (OB) women vs. normal weight (NW) women. However, total autophagic activity in these placentas appeared to be decreased as it showed an increase in SQSTM1/p62 and a decrease in lysosomal biogenesis. A mouse model with a targeted deletion of the essential autophagy gene Atg7 in placental tissue showed significant placental abnormalities comparable to those seen in human placenta with maternal obesity. These included a decrease in expression of mitochondrial genes and antioxidants, and decreased lysosomal biogenesis. Strikingly, the knockout mice were developmentally programmed as they showed an increased sensitivity to high-fat diet-induced obesity, hyperglycemia, hyperinsulinemia, increased adiposity, and cardiac remodeling. In summary, our results indicate a sexual dimorphism in placental autophagy in response to maternal obesity. We also show that autophagy plays an important role in placental function and that inhibition of placental autophagy programs the offspring to obesity, and to metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sribalasubashini Muralimanoharan
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Xiaoli Gao
- b The Metabolomics Core Facility, Institutional Mass Spectrometry Laboratory, University of Texas Health Science Center , San Antonio , TX , USA
| | - Susan Weintraub
- b The Metabolomics Core Facility, Institutional Mass Spectrometry Laboratory, University of Texas Health Science Center , San Antonio , TX , USA
| | - Leslie Myatt
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA.,c Department of Ob/Gyn , Oregon Health and Science University , Portland , OR , USA
| | - Alina Maloyan
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA.,d Knight Cardiovascular Institute, Oregon Health and Science University , Portland , OR , USA
| |
Collapse
|
40
|
Myatt L, Thornburg KL. Effects of Prenatal Nutrition and the Role of the Placenta in Health and Disease. Methods Mol Biol 2018; 1735:19-46. [PMID: 29380305 DOI: 10.1007/978-1-4939-7614-0_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epidemiologic studies identified the linkage between exposures to stresses, including the type and plane of nutrition in utero with development of disease in later life. Given the critical roles of the placenta in mediating transport of nutrients between the mother and fetus and regulation of maternal metabolism, recent attention has focused on the role of the placenta in mediating the effect of altered nutritional exposures on the development of disease in later life. In this chapter we describe the mechanisms of nutrient transport in the placenta, the influence of placental metabolism on this, and how placental energetics influence placental function in response to a variety of stressors. Further the recent "recognition" that the placenta itself has a sex which affects its function may begin to help elucidate the mechanisms underlying the well-known dimorphism in development of disease in adult life.
Collapse
Affiliation(s)
- Leslie Myatt
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA. .,Bob and Charlee Moore Institute for Nutrition & Wellness, Oregon Health & Science University, Portland, OR, USA.
| | - Kent L Thornburg
- Bob and Charlee Moore Institute for Nutrition & Wellness, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
41
|
A simple method to assess group difference in RT-qPCR reference gene selection using GeNorm: The case of the placental sex. Sci Rep 2017; 7:16923. [PMID: 29209038 PMCID: PMC5717228 DOI: 10.1038/s41598-017-16916-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/20/2017] [Indexed: 01/12/2023] Open
Abstract
Normalization with proper reference genes is a crucial step in obtaining accurate mRNA expression levels in RT-qPCR experiments. GeNorm and NormFinder are two commonly used software packages that help in selecting the best reference genes, based on their expression stability. However, GeNorm does not take into account a group variable, such as sample sex, in its calculation. We demonstrate a simple calculation step to assess the variability of such parameters by multiplying the GeNorm M value with the difference of Cq values between groups. To test this, we used 28 reference gene candidates, to analyze 20 placental samples (10 of each sex), and by using HPRT1 (lower Cq values in male placentas (P = 0.017)), as a target gene. Our calculation demonstrates that the RPL30 – GAPDH reference gene combination is the better option to assess small placental sex differences in mRNA level, versus the selection obtained from GeNorm or NormFinder. The HPRT1 normalized mRNA expression level is different between placental sexes, using RPL30 and GAPDH as reference genes (P = 0.01), but not when using genes suggested by GeNorm or NormFinder. These results indicate that the proposed calculation is appropriate to assess small variations in mRNA expression between 2 groups.
Collapse
|
42
|
Pérez-Cerezales S, Ramos-Ibeas P, Rizos D, Lonergan P, Bermejo-Alvarez P, Gutiérrez-Adán A. Early sex-dependent differences in response to environmental stress. Reproduction 2017; 155:R39-R51. [PMID: 29030490 DOI: 10.1530/rep-17-0466] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022]
Abstract
Developmental plasticity enables the appearance of long-term effects in offspring caused by exposure to environmental stressors during embryonic and foetal life. These long-term effects can be traced to pre- and post-implantation development, and in both cases, the effects are usually sex specific. During preimplantation development, male and female embryos exhibit an extensive transcriptional dimorphism mainly driven by incomplete X chromosome inactivation. These early developmental stages are crucial for the establishment of epigenetic marks that will be conserved throughout development, making it a particularly susceptible period for the appearance of long-term epigenetic-based phenotypes. Later in development, gonadal formation generates hormonal differences between the sexes, and male and female placentae exhibit different responses to environmental stressors. The maternal environment, including hormones and environmental insults during pregnancy, contributes to sex-specific placental development that controls genetic and epigenetic programming during foetal development, regulating sex-specific differences, including sex-specific epigenetic responses to environmental hazards, leading to long-term effects. This review summarizes several human and animal studies examining sex-specific responses to environmental stressors during both the periconception period (caused by differences in sex chromosome dosage) and placental development (caused by both sex chromosomes and hormones). The identification of relevant sex-dependent trajectories caused by sex chromosomes and/or sex hormones is essential to define diagnostic markers and prevention/intervention protocols.
Collapse
Affiliation(s)
| | | | | | - Pat Lonergan
- School of Agriculture and Food ScienceUniversity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
43
|
Howland MA, Sandman CA, Glynn LM. Developmental origins of the human hypothalamic-pituitary-adrenal axis. Expert Rev Endocrinol Metab 2017; 12:321-339. [PMID: 30058893 PMCID: PMC6334849 DOI: 10.1080/17446651.2017.1356222] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The developmental origins of disease or fetal programming model predicts that intrauterine exposures have life long consequences for physical and psychological health. Prenatal programming of the fetal hypothalamic-pituitary-adrenal (HPA) axis is proposed as a primary mechanism by which early experiences are linked to later disease risk. Areas covered: This review describes the development of the fetal HPA axis, which is determined by an intricately timed cascade of endocrine events during gestation and is regulated by an integrated maternal-placental-fetal steroidogenic unit. Mechanisms by which stress-induced elevations in hormones of maternal, fetal, or placental origin influence the structure and function of the emerging fetal HPA axis are discussed. Recent prospective studies documenting persisting associations between prenatal stress exposures and altered postnatal HPA axis function are summarized, with effects observed beginning in infancy into adulthood. Expert commentary: The results of these studies are synthesized, and potential moderating factors are discussed. Promising areas of further research highlighted include epigenetic mechanisms and interactions between pre and postnatal influences.
Collapse
Affiliation(s)
- Mariann A. Howland
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Laura M. Glynn
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
- Department of Psychology, Chapman University, Orange, CA, USA
| |
Collapse
|
44
|
Eksteen M, Heide G, Tiller H, Zhou Y, Nedberg NH, Martinez-Zubiaurre I, Husebekk A, Skogen BR, Stuge TB, Kjær M. Anti-human platelet antigen (HPA)-1a antibodies may affect trophoblast functions crucial for placental development: a laboratory study using an in vitro model. Reprod Biol Endocrinol 2017; 15:28. [PMID: 28427432 PMCID: PMC5399428 DOI: 10.1186/s12958-017-0245-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 04/02/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a bleeding disorder caused by maternal antibodies against paternal human platelet antigens (HPAs) on fetal platelets. Antibodies against HPA-1a are accountable for the majority of FNAIT cases. We have previously shown that high levels of maternal anti-HPA-1a antibodies are associated with clinically significant reduced birth weight in newborn boys. Chronic inflammatory placental lesions are associated with increased risk of reduced birth weight and have previously been reported in connection with FNAIT pregnancies. The HPA-1a epitope is located on integrin β3 that is associated with integrin αIIb (the fibrinogen receptor) on platelets and megakaryocytes. Integrin β3 is also associated with integrin αV forming the αVβ3 integrin heterodimer, the vitronectin receptor, which is expressed on various cell types, including trophoblast cells. It is therefore thinkable that maternal anti-HPA-1a antibodies present during early pregnancy may affect placenta function through binding to the HPA-1a antigen epitope on invasive throphoblasts. The aim of the study was to examine whether interaction of a human anti-HPA-1a monoclonal antibody (mAb) with HPA-1a on trophoblast cells affect adhesion, migration and invasion of extravillous trophoblast cells. METHODS An in vitro model with human anti-HPA-1a mAb, clone 26.4, and the first trimester extravillous trophoblast cell line HTR8/SVneo was employed. The xCELLigence system was utilized to assess the possible effect of anti-HPA-1a mAb on adhesion and migration of HTR8/SVneo cells. Specially designed chambers precoated with Matrigel were used to assess the effect on the invasive capacity of cells. RESULTS We found that human anti-HPA-1a mAb 26.4 partially inhibits adhesion and migratory capacity of HTR8/SVneo cells. CONCLUSIONS Our findings suggest that anti-HPA-1a antibodies may affect trophoblast functions crucial for normal placental development. Future studies including primary throphoblast cells and polyclonal anti-HPA-1a antibodies are needed to confirm these results.
Collapse
Affiliation(s)
- Mariana Eksteen
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Gøril Heide
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Heidi Tiller
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Obstetrics and Gynecology, University Hospital of North Norway, Tromsø, Norway
| | - Yan Zhou
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San-Francisco, CA USA
| | - Nora Hersoug Nedberg
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- Prophylix Pharma AS, Tromsø, Norway
| | - Inigo Martinez-Zubiaurre
- Bone and Joint research group, Department of Clinical Medicine, UiT- The Arctic University of Norway, Tromsø, Norway
| | - Anne Husebekk
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Bjørn R. Skogen
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Tor B. Stuge
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Mette Kjær
- Immunology research group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
45
|
Meakin AS, Saif Z, Jones AR, Aviles PFV, Clifton VL. Review: Placental adaptations to the presence of maternal asthma during pregnancy. Placenta 2017; 54:17-23. [PMID: 28131319 DOI: 10.1016/j.placenta.2017.01.123] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/10/2017] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
Asthma is a highly prevalent chronic medical condition affecting an estimated 12% of pregnant, women each year, with prevalence of asthma greatest (up to 16%) among the socially disadvantaged. Maternal asthma is associated with significant perinatal morbidity and mortality including preterm births, neonatal hospitalisations and low birthweight outcomes each year. We have identified that the placenta adapts to the presence of chronic, maternal asthma during pregnancy in a sex specific manner that may confer sex differences in fetal outcome. The male fetus was at greater risk of a poor outcome than a female fetus in the presence of maternal asthma and an acute inflammatory event such as an asthma exacerbation. This review will examine the role of sex specific differences in placental function on fetal growth and survival.
Collapse
Affiliation(s)
- A S Meakin
- Mater Medical Research Institute, University of Queensland, Brisbane, Australia
| | - Z Saif
- Mater Medical Research Institute, University of Queensland, Brisbane, Australia
| | - A R Jones
- Mater Medical Research Institute, University of Queensland, Brisbane, Australia
| | | | - V L Clifton
- Mater Medical Research Institute, University of Queensland, Brisbane, Australia.
| |
Collapse
|
46
|
Sex differences in early-life programming of the hypothalamic-pituitary-adrenal axis in humans suggest increased vulnerability in females: a systematic review. J Dev Orig Health Dis 2017; 8:244-255. [PMID: 28103963 DOI: 10.1017/s204017441600074x] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fetal glucocorticoid overexposure is a key mechanism linking early development with later-life disease. In humans, low birth weight associates with increased fasting cortisol, hypothalamic-pituitary-adrenal (HPA) axis reactivity, and with cardiovascular risk and cognitive decline. As there are sex differences in these adult diseases, we hypothesized that there may be sex differences in programming of the HPA axis in response to prenatal stressors. We conducted a systematic review following Meta-Analysis of Observational Studies in Epidemiology and Preferred Reporting Items for Systematic Reviews and Meta-Analysis. We searched Embase, MEDLINE and Web of Science from inception to 31 October 2016. We included studies related to sex differences, prenatal exposures and HPA axis. We excluded studies investigating specific disease states. The 23 included studies investigated the consequences of low birth weight, preterm birth and maternal stressors of asthma, psychosocial stress and glucocorticoid medications on HPA axis outcomes of placental glucocorticoid biology and offspring HPA axis function in early life and later life. Female offspring exposed to stressors had increased HPA axis reactivity compared with males. Furthermore, the female placenta increased its permeability to maternal glucocorticoids following maternal stress with changes in the expression of 11β-hydroxysteroid dehydrogenase enzymes in response to maternal glucocorticoid exposure or asthma. Among males there was some evidence of altered diurnal cortisol secretion. We conclude that although there is some evidence of male vulnerability leading to altered diurnal cortisol secretion, the female HPA axis is more vulnerable to programming, particularly in terms of its reactivity; this suggests a mechanism underlying sex differences in later-life diseases.
Collapse
|
47
|
Brunst KJ, Rosa MJ, Jara C, Lipton LR, Lee A, Coull BA, Wright RJ. Impact of Maternal Lifetime Interpersonal Trauma on Children's Asthma: Mediation Through Maternal Active Asthma During Pregnancy. Psychosom Med 2017; 79:91-100. [PMID: 27359172 PMCID: PMC5182122 DOI: 10.1097/psy.0000000000000354] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Traumatic stressors, including child abuse and/or interpersonal violence over a woman's lifecourse, can affect the health of her children. This study examines the associations between maternal lifetime interpersonal trauma (IPT) and children's asthma by age 6 years (n = 857). METHODS Pregnant women completed the Revised Conflict Tactics Scale; IPT exposure was categorized as unexposed (55%), early (childhood and/or teen years only, 25%), late (adulthood and/or index pregnancy, 7%), and chronic (early and late, 13%). Clinician-diagnosed asthma in children was reported by mothers at each follow-up visit until the child reached age 6 years. We examined the effects of maternal IPT categories and child's asthma using logistic regression. Using structural equation models, we also examined indirect relationships between maternal chronic IPT and child asthma operating through active asthma in pregnancy, prepregnancy BMI, prenatal smoking, and/or increased exposure to other adverse life events or environmental toxins prenatally. Effect modification by the child's sex was examined. RESULTS Mothers were primarily Hispanic (55%) or black (30%) with less than high school education (62%). In logistic regression models, chronic maternal IPT (compared with unexposed) was associated with asthma in boys (odds ratio = 2.87, 95% confidence interval = 1.48-5.57) but not girls (odds ratio = 0.69, 95% confidence interval = 0.23-2.12; pinteraction = .042). In structural equation models, chronic IPT was associated with maternal active asthma in pregnancy (β = 0.59, p < .001), maternal active asthma was associated with children's asthma (β = 0.20, p = .009), and the total indirect effect for this path was significant (β = 0.12, p = .031). Associations were most evident among boys. CONCLUSIONS Mothers' history of chronic IPT was associated with asthma in boys. This association was mediated through active maternal asthma in pregnancy.
Collapse
Affiliation(s)
- Kelly J. Brunst
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria José Rosa
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Calvin Jara
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lianna R. Lipton
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brent A. Coull
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - Rosalind J. Wright
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Latendresse G, Founds S. The Fascinating and Complex Role of the Placenta in Pregnancy and Fetal Well-being. J Midwifery Womens Health 2016; 60:360-70. [PMID: 26255798 DOI: 10.1111/jmwh.12344] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Existing evidence implicates the placenta as the origin of some common pregnancy complications. Moreover, some maternal conditions, such as inadequate nutrition, diabetes, and obesity, are known to adversely affect placental function, with subsequent negative impact on the fetus and newborn. The placenta may also contribute to fetal programming with health consequences into adulthood, such as cardiovascular, metabolic, and mental health disorders. There is evidence that altered placental development, specifically impaired trophoblast invasion and spiral artery remodeling in the first trimester, is the origin of preeclampsia. Prenatal care providers who understand the relationships between placental health and maternal-newborn health can better inform and guide women to optimize health early in pregnancy and prior to conception. This article reviews the current understanding of placental function; placental contributions to normal fetal brain development and timing of birth; and impact of maternal nutrition, obesity, and diabetes on the placenta.
Collapse
|
49
|
Maternal obesity alters brain derived neurotrophic factor (BDNF) signaling in the placenta in a sexually dimorphic manner. Placenta 2016; 49:55-63. [PMID: 28012455 DOI: 10.1016/j.placenta.2016.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Obesity is a major clinical problem in obstetrics being associated with adverse pregnancy outcomes and fetal programming. Brain derived neurotrophic factor (BDNF), a validated miR-210 target, is necessary for placental development, fetal growth, glucose metabolism, and energy homeostasis. Plasma BDNF levels are reduced in obese individuals; however, placental BDNF has yet to be studied in the context of maternal obesity. In this study, we investigated the effect of maternal obesity and sexual dimorphism on placental BDNF signaling. METHODS BDNF signaling was measured in placentas from lean (pre-pregnancy BMI < 25) and obese (pre-pregnancy BMI>30) women at term without medical complications that delivered via cesarean section without labor. MiRNA-210, BDNF mRNA, proBDNF, and mature BDNF were measured by RT - PCR, ELISA, and Western blot. Downstream signaling via TRKB (BDNF receptor) was measured using Western blot. RESULTS Maternal obesity was associated with increased miRNA-210 and decreased BDNF mRNA in placentas from female fetuses, and decreased proBDNF in placentas from male fetuses. We also identified decreased mature BDNF in placentas from male fetuses when compared to female fetuses. Mir-210 expression was negatively correlated with mature BDNF protein. TRKB phosphorylated at tyrosine 817, not tyrosine 515, was increased in placentas from obese women. Maternal obesity was associated with increased phosphorylation of MAPK p38 in placentas from male fetuses, but not phosphorylation of ERK p42/44. DISCUSSION BDNF regulation is complex and highly regulated. Pre-pregnancy/early maternal obesity adversely affects BDNF/TRKB signaling in the placenta in a sexually dimorphic manner. These data collectively suggest that induction of placental TRKB signaling could ameliorate the placental OB phenotype, thus improving perinatal outcome.
Collapse
|
50
|
Cantonwine DE, Meeker JD, Ferguson KK, Mukherjee B, Hauser R, McElrath TF. Urinary Concentrations of Bisphenol A and Phthalate Metabolites Measured during Pregnancy and Risk of Preeclampsia. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1651-1655. [PMID: 27177253 PMCID: PMC5047771 DOI: 10.1289/ehp188] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 12/18/2015] [Accepted: 04/21/2016] [Indexed: 05/20/2023]
Abstract
BACKGROUND Preeclampsia represents a major cause of maternal mortality and morbidity worldwide. Although it is known that the placenta plays a central role in development of preeclampsia, investigation into the contribution of environmental toxicants to the risk of preeclampsia has been sparse. OBJECTIVES In the present study we examined the relationship between longitudinally measured urinary BPA and phthalate metabolite concentrations during gestation and preeclampsia. METHODS A nested case-control study of preterm birth was performed in 2011 from women enrolled in a prospective birth cohort study at Brigham and Women's Hospital in Boston. There were 50 cases of preeclampsia as part of this study. Urine samples were analyzed for concentrations of BPA and nine phthalate metabolites several times during pregnancy. Adjusted Cox proportional hazard models were used to calculate hazard ratios of preeclampsia in association with an interquartile range increase in BPA and phthalate concentrations and were weighted to reflect results generalizable to the base population. RESULTS Adjusted hazard ratios indicated that an interquartile range increase of urinary concentrations of BPA (1.53; 95% CI: 1.04, 2.25) and MEP (monoethyl phthalate) (1.72; 95% CI: 1.28, 2.30) at 10 weeks gestation was associated with onset of preeclampsia, whereas significantly elevated hazard ratios were found across gestation for all DEHP [di(2-ethylhexyl) phthalate] metabolites. These relationships differed based on infant sex. CONCLUSIONS Urinary concentrations of BPA and several phthalate metabolites were significantly associated with increased risk of preeclampsia. If validated, these results indicate an environmental contribution of endocrine-disrupting chemicals to preeclampsia and suggest a modifiable means to reduce the mortality and morbidity associated with this condition. CITATION Cantonwine DE, Meeker JD, Ferguson KK, Mukherjee B, Hauser R, McElrath TF. 2016. Urinary concentrations of bisphenol A and phthalate metabolites measured during pregnancy and risk of preeclampsia. Environ Health Perspect 124:1651-1655; http://dx.doi.org/10.1289/EHP188.
Collapse
Affiliation(s)
- David E. Cantonwine
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Address correspondence to D.E. Cantonwine, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA. Telephone: (617) 525-9756. E-mail: mailto:
| | | | | | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Thomas F. McElrath
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|