1
|
Carver AJ, Dunnwald M, Stevens HE. A head start: The relationship of placental factors to craniofacial and brain development. Dev Dyn 2025. [PMID: 40105397 DOI: 10.1002/dvdy.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
In recent years, the importance of placental function for fetal neurodevelopment has become increasingly studied. This field, known as neuroplacentology, has greatly expanded possible etiologies of neurodevelopmental disorders by exploring the influence of placental function on brain development. It is also well-established that brain development is influenced by craniofacial morphogenesis. However, there is less focus on the impact of the placenta on craniofacial development. Recent research suggests the functional influence of placental nutrients and hormones on craniofacial skeletal growth, such as prolactin, growth hormone, insulin-like growth factor 1, vitamin D, sulfate, and calcium, impacting both craniofacial and brain development. Therefore, interactions between the placenta and both fetal neurodevelopment and craniofacial development likely influence the growth and morphology of the head as a whole. This review discusses the role of placental hormone production and nutrient delivery in the development of the fetal head-defined as craniofacial and brain tissue together-expanding on the more established focus on brain development to also include the skull (or cranium) and face.
Collapse
Affiliation(s)
- Annemarie Jenna Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Martine Dunnwald
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, Iowa City, Iowa, USA
| | - Hanna Elizabeth Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Hawk Intellectual and Developmental Disabilities Research Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Maurer SV, Hing BWQ, Lussier S, Radhakrishna S, Davis JLB, Abbott PW, Michaelson JJ, Stevens HE. Prenatal stress alters mouse offspring dorsal striatal development and placental function in sex-specific ways. J Psychiatr Res 2025; 182:149-160. [PMID: 39809011 PMCID: PMC11959308 DOI: 10.1016/j.jpsychires.2024.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Prenatal stress is a risk factor for neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD). However, how early stress modification of brain development contributes to this pathophysiology is poorly understood. Ventral forebrain regions such as dorsal striatum are of particular interest: dorsal striatum modulates movement and cognition, is altered in NDDs, and has a primarily GABAergic population. Here, we examine effects of prenatal stress on adult movement, cognition, and dorsal striatum neurobiology in mice using striatal-dependent behavioral assays, immunohistochemistry, embryonic ventral forebrain transcriptomics, and placental transcriptomics. We found prenatal stress affected adult procedural, habit, and reversal learning in sex-specific ways. Stress also increased adult dorsal striatal GABAergic neurons - an effect largely driven by males. We sought to examine the developmental origins of these adult brain changes. We found similar sex-specific dorsal striatal cellular changes in earlier points of development. The dorsal striatum primordium--embryonic ventral forebrain-showed that prenatal stress increased DNA replication and cell cycle pathways in male but not female transcriptomics and cellular biology. Unique signatures may have arisen from male-female placental differences. Stress-induced placental transcriptomics showed upregulated morphogenesis pathways in males while females downregulated morphogenic, hormonal, and cellular response pathways. Our findings suggest that prenatal stress could affect placenta function and also alter the GABAergic population of dorsal striatum differentially between the sexes.
Collapse
Affiliation(s)
- Sara V Maurer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Benjamin W Q Hing
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Stephanie Lussier
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SL is now with Moderna, USA
| | - Sreya Radhakrishna
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SR is now at Albert Einstein College of Medicine, USA
| | - Jada L B Davis
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Parker W Abbott
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
3
|
Yuan Z, Su T, Yang L, Xi L, Wang HJ, Ji Y. Maternal Glycemia and Its Pattern Associated with Offspring Neurobehavioral Development: A Chinese Birth Cohort Study. Nutrients 2025; 17:257. [PMID: 39861387 PMCID: PMC11767945 DOI: 10.3390/nu17020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study investigates the impact of maternal glycemic levels during early and late pregnancy on offspring neurodevelopment in China. METHODS Fasting plasma glucose (FPG) and triglyceride (TG) levels were measured in maternal blood during pregnancy, and the TyG index was calculated to assess insulin resistance. Hyperglycemia was defined as FPG > 5.1 mmol/L. Neurodevelopmental outcomes in offspring aged 6-36 months were evaluated using the China Developmental Scale for Children, focusing on developmental delay (DD) and developmental quotient (DQ). Mothers were categorized into four glycemic groups: healthy glycemia group (HGG), early pregnancy hyperglycemia group (EHG), late pregnancy hyperglycemia group (LHG), and full-term hyperglycemia group (FHG). Linear and logistic regression models were applied. RESULTS Among 1888 mother-child pairs, hyperglycemia and FPG were associated with an increased risk of overall DD (aOR = 1.68; 95% CI 1.07-2.64) and lower DQ (aBeta = -1.53; 95% CI -2.70 to -0.36). Elevated FPG was linked to DD in fine motor and social behaviors. Compared to HGG, LHG and FHG significantly increased the risk of overall DD (aOR = 2.18; 95% CI 1.26-3.77; aOR = 2.64; 95% CI 1.38-5.05), whereas EHG did not. Male offspring were particularly vulnerable to early pregnancy hyperglycemia (aBeta = -2.80; 95% CI -4.36 to -1.34; aOR = 2.05; 95% CI 1.10-3.80). CONCLUSIONS Maternal glycemic levels during pregnancy influence offspring neurodevelopment, with persistent hyperglycemia significantly increasing DD risk. Early pregnancy hyperglycemia particularly affects male offspring, underscoring the need for glycemic management during pregnancy.
Collapse
Affiliation(s)
- Zhichao Yuan
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Tao Su
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Li Yang
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Lei Xi
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Yuelong Ji
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
4
|
Zoubovsky SP, Muglia LJ. Transplacental signals involved in the programming effects of prenatal psychosocial stress on neurodevelopment. Neurotoxicol Teratol 2025; 107:107424. [PMID: 39755178 DOI: 10.1016/j.ntt.2025.107424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Exposure to psychosocial stress during pregnancy has been associated with the emergence of neurodevelopmental and neuropsychiatric disorders in offspring. The placenta is known to orchestrate various functions that are essential for normal fetal development, including the brain. It has therefore been postulated that alterations in such functions, and downstream signaling, have the potential to dramatically affect brain developmental trajectories and contribute to adverse neurodevelopmental outcomes. This review will focus on discussing various placental functions that have been proposed to be affected by exposure to prenatal psychosocial stress and the implications of such disruptions on long-term neurodevelopmental programming.
Collapse
Affiliation(s)
- Sandra P Zoubovsky
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Louis J Muglia
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Office of the President, Burroughs Wellcome Fund, Research Triangle Park, Durham, NC, United States.
| |
Collapse
|
5
|
Denizli M, Ropa J, Beasley L, Ghosh J, DeVanna K, Spice T, Haneline LS, Capitano M, Kua KL. Glucose intolerance as a consequence of hematopoietic stem cell dysfunction in offspring of obese mice. Mol Metab 2024; 88:102008. [PMID: 39142562 PMCID: PMC11395717 DOI: 10.1016/j.molmet.2024.102008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/22/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE Maternal obesity is increasingly common and negatively impacts offspring health. Children of mothers with obesity are at higher risk of developing diseases linked to hematopoietic system abnormalities and metabolism such as type 2 diabetes. Interestingly, disease risks are often dependent on the offspring's sex, suggesting sex-specific reprogramming effect of maternal obesity on offspring hematopoietic stem and progenitor cell (HSPC) function. However, the impact of maternal obesity exposure on offspring HSPC function, and the capability of HSPC to regulate offspring metabolic health is largely understudied. This study aims to test the hypothesis that offspring of obese mice exhibit sex-differences in HSPC function that affect offspring's metabolic health. METHODS We first assessed bone marrow hematopoietic stem and progenitor cell phenotype using postnatal day 21 (P21) and 8-week-old C57BL/6J mice born to control and diet-induced obese dams. We also sorted HSPC (Lineage-, Sca1+, cKit + cells) from P21 mice for competitive primary and secondary transplant, as well as transcriptomic analysis. Body weight, adiposity, insulin tolerance test and glucose tolerance tests were performed in primary and secondary transplant recipient animals. RESULTS We discovered sex-differences in offspring HSPC function in response to maternal obesity exposure, where male offspring of obese dams (MatOb) showed decreased HSPC numbers and engraftment, while female MatOb offspring remained largely unaffected. RNA-seq revealed immune stimulatory pathways in female MatOb offspring. Finally, only recipients of male MatOb offspring HSPC exhibited glucose intolerance. CONCLUSIONS This study demonstrated the lasting effect of maternal obesity exposure on offspring HSPC function and implicates HSPC in metabolic regulation.
Collapse
Affiliation(s)
- Merve Denizli
- Department of Pediatrics, Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Lindsay Beasley
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Kelli DeVanna
- Department of Pediatrics, Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Taylor Spice
- Department of Pediatrics, Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Laura S Haneline
- Department of Pediatrics, Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Maegan Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA.
| | - Kok Lim Kua
- Department of Pediatrics, Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, 46202, USA; Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, 46202, USA; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, 46202, USA.
| |
Collapse
|
6
|
Shah RG, Salafia CM, Girardi T, Rukat C, Brunner J, Barrett ES, O'Connor TG, Misra DP, Miller RK. Maternal affective symptoms and sleep quality have sex-specific associations with placental topography. J Affect Disord 2024; 360:62-70. [PMID: 38806063 DOI: 10.1016/j.jad.2024.05.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/10/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND The impacts of prenatal maternal affective symptoms on the placental structure are not well-established. Employing Geographic Information System (GIS) spatial autocorrelation, Moran's I, can help characterize placental thickness uniformity/variability and evaluate the impacts of maternal distress on placental topography. METHODS This study (N = 126) utilized cohort data on prenatal maternal affective symptoms and placental 2D and 3D morphology. Prenatal maternal depression, stress, anxiety and sleep quality were scored for each trimester using the Edinburgh Postnatal Depression Scale (EPDS), Stressful Life Event Scale (SLE), Penn State Worry Questionnaire (PSWQ), and Pittsburgh Sleep Quality Index (PSQI), respectively. Placental shape was divided into Voronoi cells and thickness variability among these cells was computed using Moran's I for 4-nearest neighbors and neighbors within a 10 cm radius. Sex-stratified Spearman correlations and linear regression were used to study associations between mean placental thickness, placental GIS variables, placental weight and the average score of each maternal variable. RESULTS For mothers carrying boys, poor sleep was associated with higher mean thickness (r = 0.308,p = 0.035) and lower placental thickness uniformity (r = -0.36,p = 0.012). Lower placental weight (r = 0.395,p = 0.003), higher maternal depression (r = -0.318,p = 0.019) and worry/anxiety (r = -0.362,p = 0.007) were associated with lower placental thickness uniformity for mothers carrying girls. LIMITATIONS The study is exploratory and not all GIS models were developed. Excluding high-risk pregnancies prevented investigating pregnancy complications related hypotheses. A larger sample size is needed for greater confidence for clinical application. CONCLUSIONS Placental topography can be studied using GIS theory and has shown that prenatal maternal affective symptoms and sleep have sex-specific associations with placental thickness.
Collapse
Affiliation(s)
- Ruchit G Shah
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA.
| | - Carolyn M Salafia
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Theresa Girardi
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Cate Rukat
- Placental Analytics, LLC, New Rochelle, USA and New York State Institute for Basic Research, Staten Island, USA
| | - Jessica Brunner
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health; Environmental and Occupational Health Sciences Institute, Piscataway, USA
| | - Thomas G O'Connor
- Departments of Psychiatry, Obstetrics/Gynecology, Pediatrics, University of Rochester, School of Medicine and Dentistry, Rochester, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, Michigan State University, MI, USA
| | - Richard K Miller
- Departments of Obstetrics and Gynecology, Environmental Medicine, Pathology, and Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, USA
| |
Collapse
|
7
|
Steele JW, Krishnan V, Finnell RH. Mechanisms of neurodevelopmental toxicity of topiramate. Crit Rev Toxicol 2024; 54:465-475. [PMID: 38995641 PMCID: PMC11296906 DOI: 10.1080/10408444.2024.2368552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024]
Abstract
Prescriptions for antiseizure medications (ASMs) have been rapidly growing over the last several decades due, in part, to an expanding list of clinical indications for which they are now prescribed. This trend has raised concern for potential adverse neurodevelopmental outcomes in ASM-exposed pregnancies. Recent large scale population studies have suggested that the use of topiramate (TOPAMAX, Janssen-Cilag), when prescribed for seizure control, migraines, and/or weight management, is associated with an increased risk for autism spectrum disorder (ASD), intellectual disability, and attention-deficit/hyperactivity disorder (ADHD) in exposed offspring. Here, we critically review epidemiologic evidence demonstrating the neurobehavioral teratogenicity of topiramate and speculate on the neuromolecular mechanisms by which prenatal exposure may perturb neurocognitive development. Specifically, we explore the potential role of topiramate's pharmacological interactions with ligand- and voltage-gated ion channels, especially GABAergic signaling, its effects on DNA methylation and histone acetylation, whether topiramate induces oxidative stress, and its association with fetal growth restriction as possible mechanisms contributing to neurodevelopmental toxicity. Resolving this biology will be necessary to reduce the risk of adverse pregnancy outcomes caused by topiramate or other ASMs.
Collapse
Affiliation(s)
- John W. Steele
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vaishnav Krishnan
- Departments of Neurology, Neuroscience and Psychiatry, and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard H. Finnell
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Nazzari S, Grumi S, Mambretti F, Villa M, Giorda R, Bordoni M, Pansarasa O, Borgatti R, Provenzi L. Sex-dimorphic pathways in the associations between maternal trait anxiety, infant BDNF methylation, and negative emotionality. Dev Psychopathol 2024; 36:908-918. [PMID: 36855816 DOI: 10.1017/s0954579423000172] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Maternal antenatal anxiety is an emerging risk factor for child emotional development. Both sex and epigenetic mechanisms, such as DNA methylation, may contribute to the embedding of maternal distress into emotional outcomes. Here, we investigated sex-dependent patterns in the association between antenatal maternal trait anxiety, methylation of the brain-derived neurotrophic factor gene (BDNF DNAm), and infant negative emotionality (NE). Mother-infant dyads (N = 276) were recruited at delivery. Maternal trait anxiety, as a marker of antenatal chronic stress exposure, was assessed soon after delivery using the Stait-Trait Anxiety Inventory (STAI-Y). Infants' BDNF DNAm at birth was assessed in 11 CpG sites in buccal cells whereas infants' NE was assessed at 3 (N = 225) and 6 months (N = 189) using the Infant Behavior Questionnaire-Revised (IBQ-R). Hierarchical linear analyses showed that higher maternal antenatal anxiety was associated with greater 6-month-olds' NE. Furthermore, maternal antenatal anxiety predicted greater infants' BDNF DNAm in five CpG sites in males but not in females. Higher methylation at these sites was associated with greater 3-to-6-month NE increase, independently of infants' sex. Maternal antenatal anxiety emerged as a risk factor for infant's NE. BDNF DNAm might mediate this effect in males. These results may inform the development of strategies to promote mothers and infants' emotional well-being.
Collapse
Affiliation(s)
- Sarah Nazzari
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Serena Grumi
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabiana Mambretti
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Marco Villa
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Roberto Giorda
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
9
|
Gan H, Xing Y, Tong J, Lu M, Yan S, Huang K, Wu X, Tao S, Gao H, Pan Y, Dai J, Tao F. Impact of Gestational Exposure to Individual and Combined Per- and Polyfluoroalkyl Substances on a Placental Structure and Efficiency: Findings from the Ma'anshan Birth Cohort. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:6117-6127. [PMID: 38525964 DOI: 10.1021/acs.est.3c09611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Prenatal exposure to perfluoroalkyl and polyfluoroalkyl substances (PFASs) is inevitable among pregnant women. Nevertheless, there is a scarcity of research investigating the connections between prenatal PFAS exposure and the placental structure and efficiency. Based on 712 maternal-fetal dyads in the Ma'anshan Birth Cohort, we analyzed associations between individual and mixed PFAS exposure and placental measures. We repeatedly measured 12 PFAS in the maternal serum during pregnancy. Placental weight, scaling exponent, chorionic disc area, and disc eccentricity were used as the outcome variables. Upon adjusting for confounders and implementing corrections for multiple comparisons, we identified positive associations between branched perfluorohexane sulfonate (br-PFHxS) and 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA) with placental weight. Additionally, a positive association was observed between br-PFHxS and the scaling exponent, where a higher scaling exponent signified reduced placental efficiency. Based on neonatal sex stratification, female infants were found to be more susceptible to the adverse effects of PFAS exposure. Mixed exposure modeling revealed that mixed PFAS exposure was positively associated with placental weight and scaling exponent, particularly during the second and third trimesters. Furthermore, br-PFHxS and 6:2 Cl-PFESA played major roles in the placental measures. This study provides the first epidemiological evidence of the relationship between prenatal PFAS exposure and placental measures.
Collapse
Affiliation(s)
- Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Yanan Xing
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Mengjuan Lu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan 243011 Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| | - Shuman Tao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hui Gao
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022 Anhui, China
| | - Yitao Pan
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032 Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032 Anhui, China
| |
Collapse
|
10
|
Chan JC, Alenina N, Cunningham AM, Ramakrishnan A, Shen L, Bader M, Maze I. Serotonin Transporter-dependent Histone Serotonylation in Placenta Contributes to the Neurodevelopmental Transcriptome. J Mol Biol 2024; 436:168454. [PMID: 38266980 PMCID: PMC10957302 DOI: 10.1016/j.jmb.2024.168454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
Brain development requires appropriate regulation of serotonin (5-HT) signaling from distinct tissue sources across embryogenesis. At the maternal-fetal interface, the placenta is thought to be an important contributor of offspring brain 5-HT and is critical to overall fetal health. Yet, how placental 5-HT is acquired, and the mechanisms through which 5-HT influences placental functions, are not well understood. Recently, our group identified a novel epigenetic role for 5-HT, in which 5-HT can be added to histone proteins to regulate transcription, a process called H3 serotonylation. Here, we show that H3 serotonylation undergoes dynamic regulation during placental development, corresponding to gene expression changes that are known to influence key metabolic processes. Using transgenic mice, we demonstrate that placental H3 serotonylation is dependent on 5-HT uptake by the serotonin transporter (SERT/SLC6A4). SERT deletion robustly reduces enrichment of H3 serotonylation across the placental genome, and disrupts neurodevelopmental gene networks in early embryonic brain tissues. Thus, these findings suggest a novel role for H3 serotonylation in coordinating placental transcription at the intersection of maternal physiology and offspring brain development.
Collapse
Affiliation(s)
- Jennifer C Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ashley M Cunningham
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Charité Universitätsmedizin Berlin, Berlin, Germany; Institute for Biology, University of Lübeck, Germany
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
11
|
Chan JC, Alenina N, Cunningham AM, Ramakrishnan A, Shen L, Bader M, Maze I. Serotonin transporter-dependent histone serotonylation in placenta contributes to the neurodevelopmental transcriptome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567020. [PMID: 38014301 PMCID: PMC10680709 DOI: 10.1101/2023.11.14.567020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Brain development requires appropriate regulation of serotonin (5-HT) signaling from distinct tissue sources across embryogenesis. At the maternal-fetal interface, the placenta is thought to be an important contributor of offspring brain 5-HT and is critical to overall fetal health. Yet, how placental 5-HT is acquired, and the mechanisms through which 5-HT influences placental functions, are not well understood. Recently, our group identified a novel epigenetic role for 5-HT, in which 5-HT can be added to histone proteins to regulate transcription, a process called H3 serotonylation. Here, we show that H3 serotonylation undergoes dynamic regulation during placental development, corresponding to gene expression changes that are known to influence key metabolic processes. Using transgenic mice, we demonstrate that placental H3 serotonylation largely depends on 5-HT uptake by the serotonin transporter (SERT/SLC6A4). SERT deletion robustly reduces enrichment of H3 serotonylation across the placental genome, and disrupts neurodevelopmental gene networks in early embryonic brain tissues. Thus, these findings suggest a novel role for H3 serotonylation in coordinating placental transcription at the intersection of maternal physiology and offspring brain development.
Collapse
Affiliation(s)
- Jennifer C Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ashley M Cunningham
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Germany
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
12
|
Shuffrey LC, Morales S, Jacobson MH, Enlow MB, Ghassabian A, Margolis AE, Lucchini M, Carroll KN, Crum RM, Dabelea D, Deutsch A, Fifer WP, Goldson B, Hockett CW, Mason WA, Jacobson LT, O’Connor TG, Pini N, Rayport Y, Sania A, Trasande L, Wright RJ, Lee S, Monk C. Association of Gestational Diabetes Mellitus and Perinatal Maternal Depression with Early Childhood Behavioral Problems: An Environmental Influences on Child Health Outcomes (ECHO) Study. Child Dev 2023; 94:1595-1609. [PMID: 37132048 PMCID: PMC10620104 DOI: 10.1111/cdev.13938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 03/16/2023] [Accepted: 03/31/2023] [Indexed: 05/04/2023]
Abstract
This study examined the association of gestational diabetes mellitus (GDM), prenatal, and postnatal maternal depressive symptoms with externalizing, internalizing, and autism spectrum problems on the Preschool Child Behavior Checklist in 2379 children aged 4.12 ± 0.60 (48% female; 47% White, 32% Black, 15% Mixed Race, 4% Asian, <2% American Indian/Alaskan Native, <2% Native Hawaiian; 23% Hispanic). Data were collected from the NIH Environmental influences on Child Health Outcomes (ECHO) Program from 2009-2021. GDM, prenatal, and postnatal maternal depressive symptoms were each associated with increased child externalizing and internalizing problems. GDM was associated with increased autism behaviors only among children exposed to perinatal maternal depressive symptoms above the median level. Stratified analyses revealed a relation between GDM and child outcomes in males only.
Collapse
Affiliation(s)
- Lauren C. Shuffrey
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Santiago Morales
- Department of Psychology, University of Southern California, Los Angeles, California, USA
| | - Melanie H. Jacobson
- Department of Pediatrics, Division of Environmental Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Akhgar Ghassabian
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Amy E. Margolis
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Maristella Lucchini
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Kecia N. Carroll
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rosa M. Crum
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - William P. Fifer
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute
| | - Brandon Goldson
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christine W. Hockett
- Avera Research Institute; Sioux Falls, South Dakota, USA
- Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, South Dakota, USA
| | - W. Alex Mason
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Lisette T. Jacobson
- Department of Population Health, Department of Obstetrics & Gynecology, University of Kansas School of Medicine, Wichita, Kansas, USA
| | - Thomas G O’Connor
- Departments of Psychiatry, Psychology, Neuroscience, and Obstetrics and Gynecology, University of Rochester, Rochester, New York, USA
| | - Nicolò Pini
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Yael Rayport
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Ayesha Sania
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Leonardo Trasande
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| | | | - Seonjoo Lee
- Mailman School of Public Health, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Catherine Monk
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Departments of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
- Division of Behavioral Medicine, New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
13
|
Anamthathmakula P, Shallie PD, Nayak N, Dhal S, Vivian JL, Mor G, Soares MJ, Nayak NR. Variable Cre Recombination Efficiency in Placentas of Cyp19-Cre ROSA mT/mG Transgenic Mice. Cells 2023; 12:2096. [PMID: 37626906 PMCID: PMC10453067 DOI: 10.3390/cells12162096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/06/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The aromatase-Cre recombinase (Cyp19-Cre) transgenic mouse model has been extensively used for placenta-specific gene inactivation. In a pilot study, we observed unexpected phenotypes using this mouse strain, which prompted an extensive characterization of Cyp19-Cre placental phenotypes using ROSAmT/mG transgenic reporter mice. The two strains were mated to generate bi-transgenic Cyp19-Cre;ROSAmT/mG mice following a standard transgenic breeding scheme, and placental and fetal tissues were analyzed on embryonic day 17.5. Both maternal and paternal Cre inheritance were analyzed by mating the respective Cyp19-Cre and ROSAmT/mG males and females. The genotype results showed the expected percentage of Cyp19-Cre;ROSAmT/mG fetuses (73%) and Cre mRNA was expressed in all of the Cyp19-Cre placentas. However, surprisingly, only about 50% of the Cyp19-Cre;ROSAmT/mG placentas showed Cre-mediated recombinase activity as demonstrated by placental enhanced green fluorescent protein (EGFP) expression. Further genetic excision analysis of the placentas revealed consistent results showing the absence of excision of the tdTomato in all of the Cyp19-Cre;ROSAmT/mG placentas lacking EGFP expression. Moreover, among the EGFP-expressing placentas, there was wide variability in recombination efficiency, even in placentas from the same litter, leading to a mosaic pattern of EGFP expression in different zones and cell types of the placentas. In addition, we observed a significantly higher percentage of Cre recombination activity in placentas with maternal Cre inheritance. Our results show frequent mosaicism, inconsistent recombination activity, and parent-of-origin effects in placentas from Cyp19-Cre;ROSAmT/mG mice, suggesting that tail-biopsy genotype results may not necessarily indicate the excision of floxed genes in Cyp19-Cre positive placentas. Thus, placenta-specific mutagenesis studies using the Cyp19-Cre model require extensive characterization and careful interpretation of the placental phenotypes for each floxed allele.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Philemon D. Shallie
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Neha Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Sabita Dhal
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Jay L. Vivian
- Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Nihar R. Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
14
|
Lodefalk M, Chelslín F, Patriksson Karlsson J, Hansson SR. Placental Changes and Neuropsychological Development in Children-A Systematic Review. Cells 2023; 12:cells12030435. [PMID: 36766778 PMCID: PMC9913696 DOI: 10.3390/cells12030435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Placental dysfunction may increase the offspring's later-life disease risk. The objective of this systematic review was to describe associations between pathological placental changes and neuropsychological outcomes in children after the neonatal period. The inclusion criteria were human studies; original research; direct placental variables; neuropsychological outcomes; and analysis between their associations. The exclusion criterion was the offspring's age-0-28 days or >19 years. The MEDLINE and EMBASE databases were last searched in May 2022. We utilized the ROBINS-I for the risk of bias assessment and performed a narrative synthesis. In total, 3252 studies were identified, out of which 16 were included (i.e., a total of 15,862 participants). Half of the studies were performed on children with neonatal complications, and 75% of the studies reported an association between a placental change and an outcome; however, following the completion of the funnel plots, a risk of publication bias was indicated. The largest study described a small association between placental size and a risk of psychiatric symptoms in boys only. Inconsistency between the studies limited the evidence in this review. In general, no strong evidence was found for an association between pathological placental changes and childhood neuropsychological outcomes after the neonatal period. However, the association between placental size and mental health in boys indicates a placental sexual dimorphism, thereby suggesting an increased vulnerability for male fetuses.
Collapse
Affiliation(s)
- Maria Lodefalk
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- Correspondence:
| | - Felix Chelslín
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Johanna Patriksson Karlsson
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences Lund, Lund University, 221 00 Lund, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
15
|
Alachkar A, Lee J, Asthana K, Vakil Monfared R, Chen J, Alhassen S, Samad M, Wood M, Mayer EA, Baldi P. The hidden link between circadian entropy and mental health disorders. Transl Psychiatry 2022; 12:281. [PMID: 35835742 PMCID: PMC9283542 DOI: 10.1038/s41398-022-02028-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 12/22/2022] Open
Abstract
The high overlapping nature of various features across multiple mental health disorders suggests the existence of common psychopathology factor(s) (p-factors) that mediate similar phenotypic presentations across distinct but relatable disorders. In this perspective, we argue that circadian rhythm disruption (CRD) is a common underlying p-factor that bridges across mental health disorders within their age and sex contexts. We present and analyze evidence from the literature for the critical roles circadian rhythmicity plays in regulating mental, emotional, and behavioral functions throughout the lifespan. A review of the literature shows that coarse CRD, such as sleep disruption, is prevalent in all mental health disorders at the level of etiological and pathophysiological mechanisms and clinical phenotypical manifestations. Finally, we discuss the subtle interplay of CRD with sex in relation to these disorders across different stages of life. Our perspective highlights the need to shift investigations towards molecular levels, for instance, by using spatiotemporal circadian "omic" studies in animal models to identify the complex and causal relationships between CRD and mental health disorders.
Collapse
Affiliation(s)
- Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA. .,Institute for Genomics and Bioinformatics, University of California, Irvine, CA, USA. .,Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA.
| | - Justine Lee
- grid.266093.80000 0001 0668 7243Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA USA
| | - Kalyani Asthana
- grid.266093.80000 0001 0668 7243Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA USA
| | - Roudabeh Vakil Monfared
- grid.266093.80000 0001 0668 7243Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA USA
| | - Jiaqi Chen
- grid.266093.80000 0001 0668 7243Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA USA
| | - Sammy Alhassen
- grid.266093.80000 0001 0668 7243Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA USA
| | - Muntaha Samad
- grid.266093.80000 0001 0668 7243Institute for Genomics and Bioinformatics, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA USA
| | - Marcelo Wood
- grid.266093.80000 0001 0668 7243Institute for Genomics and Bioinformatics, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA USA
| | - Emeran A. Mayer
- grid.266093.80000 0001 0668 7243Institute for Genomics and Bioinformatics, University of California, Irvine, CA USA ,grid.19006.3e0000 0000 9632 6718G. Oppenheimer Center of Neurobiology of Stress & Resilience and Goldman Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, CA USA
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, University of California, Irvine, CA, USA. .,Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA. .,Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
16
|
Placental Insulin Receptor Transiently Regulates Glucose Homeostasis in the Adult Mouse Offspring of Multiparous Dams. Biomedicines 2022; 10:biomedicines10030575. [PMID: 35327377 PMCID: PMC8945682 DOI: 10.3390/biomedicines10030575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
In pregnancies complicated by maternal obesity and gestational diabetes mellitus, there is strong evidence to suggest that the insulin signaling pathway in the placenta may be impaired. This may have potential effects on the programming of the metabolic health in the offspring; however, a direct link between the placental insulin signaling pathway and the offspring health remains unknown. Here, we aimed to understand whether specific placental loss of the insulin receptor (InsR) has a lasting effect on the offspring health in mice. Obesity and glucose homeostasis were assessed in the adult mouse offspring on a normal chow diet (NCD) followed by a high-fat diet (HFD) challenge. Compared to their littermate controls, InsR KOplacenta offspring were born with normal body weight and pancreatic β-cell mass. Adult InsR KOplacenta mice exhibited normal glucose homeostasis on an NCD. Interestingly, under a HFD challenge, adult male InsR KOplacenta offspring demonstrated lower body weight and a mildly improved glucose homeostasis associated with parity. Together, our data show that placenta-specific insulin receptor deletion does not adversely affect offspring glucose homeostasis during adulthood. Rather, there may potentially be a mild and transient protective effect in the mouse offspring of multiparous dams under the condition of a diet-induced obesogenic challenge.
Collapse
|
17
|
Abstract
During evolution, the cerebral cortex advances by increasing in surface and the introduction of new cytoarchitectonic areas among which the prefrontal cortex (PFC) is considered to be the substrate of highest cognitive functions. Although neurons of the PFC are generated before birth, the differentiation of its neurons and development of synaptic connections in humans extend to the 3rd decade of life. During this period, synapses as well as neurotransmitter systems including their receptors and transporters, are initially overproduced followed by selective elimination. Advanced methods applied to human and animal models, enable investigation of the cellular mechanisms and role of specific genes, non-coding regulatory elements and signaling molecules in control of prefrontal neuronal production and phenotypic fate, as well as neuronal migration to establish layering of the PFC. Likewise, various genetic approaches in combination with functional assays and immunohistochemical and imaging methods reveal roles of neurotransmitter systems during maturation of the PFC. Disruption, or even a slight slowing of the rate of neuronal production, migration and synaptogenesis by genetic or environmental factors, can induce gross as well as subtle changes that eventually can lead to cognitive impairment. An understanding of the development and evolution of the PFC provide insight into the pathogenesis and treatment of congenital neuropsychiatric diseases as well as idiopathic developmental disorders that cause intellectual disabilities.
Collapse
Affiliation(s)
- Sharon M Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
18
|
Redline RW. Placental pathology: Pathways leading to or associated with perinatal brain injury in experimental neurology, special issue: Placental mediated mechanisms of perinatal brain injury. Exp Neurol 2021; 347:113917. [PMID: 34748755 DOI: 10.1016/j.expneurol.2021.113917] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/04/2022]
Abstract
Perinatal brain injury is a multifactorial process. In utero placental physiology plays a major role in neuroprotection and the normal development of the fetal central nervous system. Advances in placental pathology have clarified several specific mechanisms of injury and the histologic lesions most strongly associated with them. This review provides an updated summary of the relevant placental anatomy and physiology, the specific placental pathways leading to brain injury, the revised Amsterdam classification system for placental pathology, and the known associations of specific placental lesions with subtypes of adverse neurologic outcomes.
Collapse
Affiliation(s)
- Raymond W Redline
- Case Western Reserve University School of Medicine, Departments of Pathology and Reproductive Biology, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106, United States of America.
| |
Collapse
|
19
|
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
|
20
|
Pang TY, Yaeger JDW, Summers CH, Mitra R. Cardinal role of the environment in stress induced changes across life stages and generations. Neurosci Biobehav Rev 2021; 124:137-150. [PMID: 33549740 PMCID: PMC9286069 DOI: 10.1016/j.neubiorev.2021.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/20/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022]
Abstract
The stress response in rodents and humans is exquisitely dependent on the environmental context. The interactive element of the environment is typically studied by creating laboratory models of stress-induced plasticity manifested in behavior or the underlying neuroendocrine mediators of the behavior. Here, we discuss three representative sets of studies where the role of the environment in mediating stress sensitivity or stress resilience is considered across varying windows of time. Collectively, these studies testify that environmental variation at an earlier time point modifies the relationship between stressor and stress response at a later stage. The metaplastic effects of the environment on the stress response remain possible across various endpoints, including behavior, neuroendocrine regulation, region-specific neural plasticity, and regulation of receptors. The timescale of such variation spans adulthood, across stages of life history and generational boundaries. Thus, environmental variables are powerful determinants of the observed diversity in stress response. The predominant role of the environment suggests that it is possible to promote stress resilience through purposeful modification of the environment.
Collapse
Affiliation(s)
- Terence Y Pang
- Florey Institute of Neuroscience and Mental Health, Parkville, 3052, VIC, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, 3010, VIC, Australia
| | - Jazmine D W Yaeger
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, 57105, USA
| | - Cliff H Summers
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, 57105, USA
| | - Rupshi Mitra
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
21
|
Wilson HA, Creighton C, Scharfman H, Choleris E, MacLusky NJ. Endocrine Insights into the Pathophysiology of Autism Spectrum Disorder. Neuroscientist 2020; 27:650-667. [PMID: 32912048 DOI: 10.1177/1073858420952046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders that affects males more frequently than females. Numerous genetic and environmental risk factors have been suggested to contribute to the development of ASD. However, no one factor can adequately explain either the frequency of the disorder or the male bias in its prevalence. Gonadal, thyroid, and glucocorticoid hormones all contribute to normal development of the brain, hence perturbations in either their patterns of secretion or their actions may constitute risk factors for ASD. Environmental factors may contribute to ASD etiology by influencing the development of neuroendocrine and neuroimmune systems during early life. Emerging evidence suggests that the placenta may be particularly important as a mediator of the actions of environmental and endocrine risk factors on the developing brain, with the male being particularly sensitive to these effects. Understanding how various risk factors integrate to influence neural development may facilitate a clearer understanding of the etiology of ASD.
Collapse
Affiliation(s)
- Hayley A Wilson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.,Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Carolyn Creighton
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Helen Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY, USA.,Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Neil J MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
22
|
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
|
23
|
Sousa FJ, Correia RG, Cruz AF, Martins JM, Rodrigues MS, Gomes CA, Ambrósio AF, Baptista FI. Sex differences in offspring neurodevelopment, cognitive performance and microglia morphology associated with maternal diabetes: Putative targets for insulin therapy. Brain Behav Immun Health 2020; 5:100075. [PMID: 34589855 PMCID: PMC8474564 DOI: 10.1016/j.bbih.2020.100075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022] Open
Abstract
Diabetes during pregnancy has been shown to affect the central nervous system (CNS) of the offspring, resulting in short- and long-term adverse effects. Children of diabetic mothers are more likely to develop cognitive impairment, also having increased susceptibility to psychiatric disorders. Microglia, the immune cells of the CNS, work as sensors of environmental changes, namely metabolic challenges, as early as the intrauterine period. During this period, microglia is actively involved in processes of neurogenesis, synaptic pruning and detection of any environmental alteration that may impact brain development. The remarkable sex dimorphism in neurodevelopment, as well as sex differences in the morphology and immune function of microglia during development, led us to clarify if maternal diabetes affects specific behavioral traits and microglia morphology during infancy in a sex-specific manner. Another important goal of this study was to clarify if insulin, the gold standard treatment of diabetes during gestation, could prevent maternal diabetes-induced behavioral changes, as well as microglia morphology, also considering sex specificities. Other molecular and cellular players potentially involved in the link between changes in metabolism and behavior were also analyzed in the hippocampus, a brain region implicated in cognition and other behavioral outcomes. Diabetes during pregnancy globally delayed female and male offspring development and was associated with impairments in recognition memory, but only in female offspring. In line with these results, at early and late infancy, some molecular and cellular markers were altered in offspring hippocampus in a sex-specific manner. The strict control of glycemia by insulin during pregnancy prevented most of the negative effects induced by uncontrolled hyperglycemia. Notably, insulin administration to diabetic dams may also modulate offspring development in a way that differs from what is observed in physiological conditions, since it promoted the expedited acquisition of developmental milestones and of discrimination ability at memory test, also inducing a hyper-ramification of male and female hippocampal microglia. Importantly, this study highlights the importance of analyzing the impact of maternal diabetes and insulin therapy, taking into account sex differences, since male and female present different vulnerabilities to hyperglycemia in this critical period of life.
Collapse
Key Words
- CA, cornu ammonis
- CTRL, offspring of control dams
- EPM, elevated plus maze
- GD, gestational day
- Insulin therapy
- Maternal diabetes
- Microglia
- NOR, novel object recognition
- Neurodevelopment
- OPF, open field
- P, postnatal day
- Recognition memory
- SEM, standard error of the mean
- STZ, offspring of streptozotocin-induced diabetic dams
- STZ + INS, offspring of insulin treated-diabetic dams
- Sex differences
Collapse
Affiliation(s)
- Fábio J Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Raquel G Correia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Alexandra F Cruz
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Joana M Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Matilde S Rodrigues
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Catarina A Gomes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| |
Collapse
|
24
|
Rosenfeld CS. The placenta-brain-axis. J Neurosci Res 2020; 99:271-283. [PMID: 32108381 DOI: 10.1002/jnr.24603] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/25/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
All mammalian species depend on the placenta, a transient organ, for exchange of gases, nutrients, and waste between the mother and conceptus. Besides serving as a conduit for such exchanges, the placenta produces hormones and other factors that influence maternal physiology and fetal development. To meet all of these adaptations, the placenta has evolved to become the most structurally diverse organ within all mammalian taxa. However, commonalities exist as to how placental responses promote survival against in utero threats and can alter the trajectory of fetal development, in particular the brain. Increasing evidence suggests that reactions of the placenta to various in utero stressors may lead to long-standing health outcomes, otherwise considered developmental origin of health and disease effects. Besides transferring nutrients and gases, the placenta produces neurotransmitters, including serotonin, dopamine, norepinephrine/epinephrine, that may circulate and influence brain development. Neurobehavioral disorders, such as autism spectrum disorders, likely trace their origins back to placental disturbances. This intimate relationship between the placenta and brain has led to coinage of the term, the placenta-brain-axis. This axis will be the focus herein, including how conceptus sex might influence it, and technologies employed to parse out the effects of placental-specific transcript expression changes on later neurobehavioral disorders. Ultimately, the placenta might provide a historical record of in utero threats the fetus confronted and a roadmap to understand how placenta responses to such encounters impacts the placental-brain-axis. Improved early diagnostic and preventative approaches may thereby be designed to mitigate such placental disruptions.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,MU Informatics Institute, University of Missouri, Columbia, MO, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, USA.,Genetics Area Program, University of Missouri, Columbia, MO, USA
| |
Collapse
|
25
|
Kratimenos P, Penn AA. Placental programming of neuropsychiatric disease. Pediatr Res 2019; 86:157-164. [PMID: 31003234 PMCID: PMC11906117 DOI: 10.1038/s41390-019-0405-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022]
Abstract
The placenta is vital for fetal growth, and compromised function is associated with abnormal development, especially of the brain. Linking placental function to brain development is a new field we have dubbed neuroplacentology. Approximately 380,000 infants in the United States each year abruptly lose placental support upon premature birth, and more than 10% of pregnancies are affected by more insidious placental dysfunction such as preeclampsia or infection. Abnormal fetal brain development or injury can lead to life-long neurological impairments, including psychiatric disorders. The majority of research connecting placental compromise to fetal brain injury has focused on gas exchange or nutritional programming, neglecting the placenta's essential neuroendocrine role. We will review the current evidence that placental dysfunction, particularly endocrine dysfunction, secretion of pro-inflammatory cytokines, or barrier breakdown may place many thousands of fetuses at risk for life-long neurodevelopmental impairments each year. Understanding how specific placental factors shape brain development and increase the risk for later psychiatric disorders, including autism, attention deficit disorder, and schizophrenia, paves the way for novel treatment strategies to maintain the normal developmental milieu and protect from further injury.
Collapse
Affiliation(s)
- Panagiotis Kratimenos
- Dept. of Pediatrics, George Washington University, Washington, DC, USA
- Neonatology, Children's National Health System, Washington, DC, USA
- Center for Neuroscience Research, Children's Research Institute at Children's National Health System, Washington, DC, USA
| | - Anna A Penn
- Dept. of Pediatrics, George Washington University, Washington, DC, USA.
- Neonatology, Children's National Health System, Washington, DC, USA.
- Center for Neuroscience Research, Children's Research Institute at Children's National Health System, Washington, DC, USA.
- Fetal and Transitional Medicine, Children's National Health System, Washington, DC, USA.
| |
Collapse
|
26
|
Valent D, Yeste N, Hernández-Castellano LE, Arroyo L, Wu W, García-Contreras C, Vázquez-Gómez M, González-Bulnes A, Bendixen E, Bassols A. SWATH-MS quantitative proteomic investigation of intrauterine growth restriction in a porcine model reveals sex differences in hippocampus development. J Proteomics 2019; 204:103391. [PMID: 31129268 DOI: 10.1016/j.jprot.2019.103391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/12/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023]
Abstract
Intrauterine growth restriction (IUGR) is characterized by reduced growth and weight of the foetus, mainly due to the lack of nutrients and oxygen. Animals affected by IUGR show changes in specific brain areas and several neuronal processes. Female offspring affected by IUGR show increased survival and development compared to males. The objective of this study was to analyse changes in the hippocampus proteome in male and female piglets affected by IUGR. Seven pregnant Iberian sows were fed from Day 35 of pregnancy onwards at 50% of their requirements. At Day 100 of pregnancy, foetuses were obtained and classified by sex and weight, as mild IUGR (Normal Body Weight) versus severe IUGR (Low Body Weight). Hippocampi were dissected and the proteomes analysed by SWATH-MS DIA. In this study, 1497 proteins were identified of which 260 were quantitatively analysed. All differential proteins were more abundant in females versus males and were involved in protein synthesis, neuronal development, metabolism, antiapoptotic signalling and vesicular transport. Our findings support that female foetuses tolerate nutrient limitation better than males, especially under mild IUGR. Under severe IUGR, females still seems to maintain normal lipid metabolism and antiapoptotic signalling, which may be related to the increased female survival. SIGNIFICANCE: In the last years, proteomics have been used to evidence differences related to sex in non-reproductive organs. Intrauterine Growth Restriction (IUGR) can affect female and male offspring differently. Female offspring has stronger protective strategies compared to males, enhancing growth and postnatal survival. Most studies regarding this issue have focused on metabolic organs (i.e. liver). However, the predominance of neurodevelopmental disorders in males suggests that the central nervous system in female offspring adapt better to nutritional stress conditions than that of males. Based on the differential protein expression in hippocampal samples, our work demonstrates that female foetuses indeed adapt better to IUGR than males, especially under mild IUGR conditions. In severe IUGR conditions, differences between males and females were not so evident, but even in this case, the remaining differences suggest increased survival in females than in males.
Collapse
Affiliation(s)
- Daniel Valent
- Departament de Bioquímica i Biologia Molecular, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Natalia Yeste
- Departament de Bioquímica i Biologia Molecular, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Lorenzo E Hernández-Castellano
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; Department of Animal Science, AU-Foulum, Aarhus University, 8830 Tjele, Denmark
| | - Laura Arroyo
- Departament de Bioquímica i Biologia Molecular, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Wei Wu
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | | | - Marta Vázquez-Gómez
- Faculty of Veterinary Sciences, UCM, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Antonio González-Bulnes
- Comparative Physiology Group, INIA, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain; Faculty of Veterinary Sciences, UCM, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Anna Bassols
- Departament de Bioquímica i Biologia Molecular, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain.
| |
Collapse
|
27
|
Liu R, Shelton RC, Eldred-Skemp N, Goldsmith J, Suglia SF. Early Exposure to Cumulative Social Risk and Trajectories of Body Mass Index in Childhood. Child Obes 2019; 15:48-55. [PMID: 30362818 PMCID: PMC6338568 DOI: 10.1089/chi.2018.0116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Childhood social risk has been associated with increased risk of childhood obesity. However, little is known about early exposure of cumulative social risk on BMI percentile (BMIp) trajectories in early childhood. METHODS Public data from the Fragile Families and Child Wellbeing Study were analyzed (N = 3809). Maternal reports of experiences of multiple social risk factors were obtained at age 1 and 3 assessments of children. Two cumulative social risk scores were calculated by summing social factors assessed at age 1 and at age 3. Child BMIp was assessed at ages 3, 5, and 9. Linear mixed models were used to examine the effect of cumulative social risk on sex-specific BMIp trajectories. RESULTS Compared with girls experiencing low social risk at either age 1 or 3, girls experiencing high social risk (≥ 2 factors) at age 1 or 3 only had higher initial BMIp at age 3 [β0 = 5.70 (95% confidence interval, CI: 0.15-1.26) and 1.37 (95% CI: -2.25 to 4.99), respectively] and had nonsignificantly greater BMIp growth rate [β1 = 0.39 (95% CI: -0.86 to 1.63) and 0.32 (95% CI: -0.86 to 1.63)]. Girls experiencing high social risk at both ages had nonsignificantly but consistently lower BMIp [β1 = -1.24 (95% CI: -2.93 to 0.46)]. In addition, girls experiencing a sum of ≥4 risk factors at both ages had lower BMIp growth rate [β1 = -1.77 (95% CI: -3.39 to -0.15)] compared to girls experiencing no risk factor. No associations were observed among boys. CONCLUSIONS Early exposure to cumulative social risk may have long-term impact on BMIp trajectories among girls, depending on timing of exposure. Understanding the effect of cumulative social risk in different contexts, including sex, chronicity, and timing of exposure, may have practical implications for informing effective intervention to combat childhood obesity.
Collapse
Affiliation(s)
- Rongzhe Liu
- Department of Epidemiology, Columbia University, New York, NY
| | - Rachel C. Shelton
- Department of Sociomedical Sciences, Columbia University, New York, NY
| | | | - Jeff Goldsmith
- Department of Biostatistics Mailman School of Public Health, Columbia University, New York, NY
| | - Shakira F. Suglia
- Department of Sociomedical Sciences, Columbia University, New York, NY.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA.,Address correspondence to: Shakira F. Suglia, ScD, Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Road, Room 4005, Atlanta, GA 30318
| |
Collapse
|
28
|
Shearer FJG, Wyrwoll CS, Holmes MC. The Role of 11β-Hydroxy Steroid Dehydrogenase Type 2 in Glucocorticoid Programming of Affective and Cognitive Behaviours. Neuroendocrinology 2019; 109:257-265. [PMID: 30884491 DOI: 10.1159/000499660] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/17/2019] [Indexed: 11/19/2022]
Abstract
Developmental exposure to stress hormones, i.e. glucocorticoids, is central to the process of prenatal programming of later-life health. Glucocorticoid overexposure, through stress or exogenous glucocorticoids, results in a reduced birthweight, as well as affective and neuropsychiatric outcomes in adults, combined with altered hypothalamus-pituitary-adrenal (HPA) axis activity. As such, glucocorticoids are tightly regulated during development through the presence of the metabolizing enzyme 11β-hydroxysteroid dehydrogenase type 2 (HSD2). HSD2 is highly expressed in 2 hubs during development, i.e. the placenta and the fetus itself, protecting the fetus from inappropriate glucocorticoid exposure early in gestation. Through manipulation of HSD2 expression in the mouse placenta and fetal tissues, we are able to determine the relative contribution of glucocorticoid exposure in each compartment. Feto-placental HSD2 deletion resulted in a reduced birthweight and the development of anxiety- and depression-like behaviours in adult mice. The placenta itself is altered by glucocorticoid overexposure, which causes reduced placental weight and vascular arborisation. Furthermore, altered flow and resistance in the umbilical vessels and modification of fetal heart function and development are observed. However, brain-specific HSD2 removal (HSD2BKO) also generated adult phenotypes of depressive-like behaviour and memory deficits, demonstrating the importance of fetal brain HSD2 expression in development. In this review we will discuss potential mechanisms underpinning early-life programming of adult neuropsychiatric disorders and the novel therapeutic potential of statins.
Collapse
Affiliation(s)
- Fraser J G Shearer
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Caitlin S Wyrwoll
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Washington, Australia
| | - Megan C Holmes
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom,
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom,
| |
Collapse
|
29
|
Misra P, Ganesh S. Sex-biased transgenerational effect of maternal stress on neurodevelopment and cognitive functions. J Genet 2018; 97:581-583. [PMID: 29932080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Piyali Misra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208 016, India.
| | | |
Collapse
|
30
|
Sex-biased transgenerational effect of maternal stress on neurodevelopment and cognitive functions. J Genet 2018. [DOI: 10.1007/s12041-018-0928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
31
|
Chan JC, Nugent BM, Bale TL. Parental Advisory: Maternal and Paternal Stress Can Impact Offspring Neurodevelopment. Biol Psychiatry 2018; 83:886-894. [PMID: 29198470 PMCID: PMC5899063 DOI: 10.1016/j.biopsych.2017.10.005] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/07/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Parental stress exposures are implicated in the risk for offspring neurodevelopmental and neuropsychiatric disorders, prompting critical examination of preconception and prenatal periods as vulnerable to environmental insults such as stress. Evidence from human studies and animal models demonstrates the influence that both maternal and paternal stress exposures have in changing the course of offspring brain development. Mechanistic examination of modes of intergenerational transmission of exposure during pregnancy has pointed to alterations in placental signaling, including changes in inflammatory, nutrient-sensing, and epigenetic pathways. Transmission of preconception paternal stress exposure is associated with changes in epigenetic marks in sperm, with a primary focus on the reprogramming of DNA methylation, histone posttranslational modifications, and small noncoding RNAs. In this review, we discuss evidence supporting the important contribution of intergenerational parental stress in offspring neurodevelopment and disease risk, and the currently known epigenetic mechanisms underlying this transmission.
Collapse
Affiliation(s)
- Jennifer C Chan
- Department of Biomedical Sciences, School of Veterinary Medicine and Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bridget M Nugent
- Department of Pharmacology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland
| | - Tracy L Bale
- Department of Pharmacology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland.
| |
Collapse
|
32
|
Zhu C, Han TL, Zhao Y, Zhou X, Mao X, Qi H, Baker PN, Zhang H. A mouse model of pre-pregnancy maternal obesity combined with offspring exposure to a high-fat diet resulted in cognitive impairment in male offspring. Exp Cell Res 2018; 368:159-166. [PMID: 29698637 DOI: 10.1016/j.yexcr.2018.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cognitive impairment is a brain dysfunction characterized by neuropsychological deficits in attention, working memory, and executive function. Maternal obesity and consumption of a high-fat diet (HFD) in the offspring has been suggested to have detrimental consequences for offspring cognitive function through its effect on the hippocampus and prefrontal cortex. Therefore, our study aimed to investigate the effects of maternal obesity and offspring HFD exposure on the brain metabolome of the offspring. METHODS In our pilot study, a LepRdb/+ mouse model was used to model pre-pregnancy maternal obesity and the c57bl/6 wildtype was used as a control group. Offspring were fed either a HFD or a low-fat control diet (LFD) after weaning (between 8 and 10 weeks). The Mirrors water maze was performed between 28 and 30 weeks to measure cognitive function. Fatty acid metabolomic profiles of the prefrontal cortex and hippocampus from the offspring at 30-32 weeks were analyzed using gas chromatography-mass spectrometry. RESULTS The memory of male offspring from obese maternal mice, consuming a HFD post-weaning, was significantly impaired when compared to the control offspring mice. No significant differences were observed in female offspring. In male mice, the fatty acid metabolites in the prefrontal cortex were most affected by maternal obesity, whereas, the fatty acid metabolites in the hippocampus were most affected by the offspring's diet. Hexadecanoic acid and octadecanoic acid were significantly affected in both the hippocampus and pre-frontal cortex, as a result of maternal obesity and a HFD in the offspring. CONCLUSION Our findings suggest that the combination of maternal obesity and HFD in the offspring can result in spatial cognitive deficiency in the male offspring, by influencing the fatty acid metabolite profiles in the prefrontal cortex and hippocampus. Further research is needed to validate the results of our pilot study.
Collapse
Affiliation(s)
- Chen Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Department of Obstetrics and Gynecology, Xin Qiao Hospital, The Second Medical College of Army Medical University, Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yalan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xiaobo Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xun Mao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Philip N Baker
- Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Liggins Institute, University of Auckland, Auckland, New Zealand; College of Medicine, Biological Sciences and Psychology, University of Leicester, UK
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
33
|
Social Origins of Developmental Risk for Mental and Physical Illness. J Neurosci 2017; 37:10783-10791. [PMID: 29118206 DOI: 10.1523/jneurosci.1822-17.2017] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Adversity in early childhood exerts an enduring impact on mental and physical health, academic achievement, lifetime productivity, and the probability of interfacing with the criminal justice system. More science is needed to understand how the brain is affected by early life stress (ELS), which produces excessive activation of stress response systems broadly throughout the child's body (toxic stress). Our research examines the importance of sex, timing and type of stress exposure, and critical periods for intervention in various brain systems across species. Neglect (the absence of sensitive and responsive caregiving) or disrupted interaction with offspring induces robust, lasting consequences in mice, monkeys, and humans. Complementary assessment of internalizing disorders and brain imaging in children suggests that early adversity can interfere with white matter development in key brain regions, which may increase risk for emotional difficulties in the long term. Neural circuits that are most plastic during ELS exposure in monkeys sustain the greatest change in gene expression, offering a mechanism whereby stress timing might lead to markedly different long-term behaviors. Rodent models reveal that disrupted maternal-infant interactions yield metabolic and behavioral outcomes often differing by sex. Moreover, ELS may further accelerate or delay critical periods of development, which reflect GABA circuit maturation, BDNF, and circadian Clock genes. Such factors are associated with several mental disorders and may contribute to a premature closure of plastic windows for intervention following ELS. Together, complementary cross-species studies are elucidating principles of adaptation to adversity in early childhood with molecular, cellular, and whole organism resolution.
Collapse
|
34
|
Chung W, Yoon S, Shin YS. Multiple exposures of sevoflurane during pregnancy induces memory impairment in young female offspring mice. Korean J Anesthesiol 2017; 70:642-647. [PMID: 29225748 PMCID: PMC5716823 DOI: 10.4097/kjae.2017.70.6.642] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/17/2017] [Accepted: 08/21/2017] [Indexed: 11/23/2022] Open
Abstract
Background Earlier studies have reported conflicting results regarding long-term behavioral consequences after anesthesia during the fetal period. Previous studies also suggest several factors that may explain such conflicting data. Thus, we examined the influence of age and sex on long-term behavioral consequences after multiple sevoflurane exposures during the fetal period. Methods C57BL/6J pregnant mice received oxygen with or without sevoflurane for 2 hours at gestational day (GD) 14-16. Offspring mice were subjected to behavioral assays for general activity (open field test), learning, and memory (fear chamber test) at postnatal day 30–35. Results Multiple sevoflurane exposures at GD 14–16 caused significant changes during the fear chamber test in young female offspring mice. Such changes did not occur in young male offspring mice. However, general activity was not affected in both male and female mice. Conclusions Multiple sevoflurane exposures in the second trimester of pregnancy affects learning and memory only in young female mice. Further studies focusing on diverse cognitive functions in an age-, sex-dependent manner may provide valuable insights regarding anesthesia-induced neurotoxicity.
Collapse
Affiliation(s)
- Woosuk Chung
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seunghwan Yoon
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Yong Sup Shin
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
35
|
Bonnin A. A Placenta-Specific Genetic Manipulation Reprograms Offspring Brain Development and Function. Biol Psychiatry 2017. [PMID: 28645358 PMCID: PMC5831337 DOI: 10.1016/j.biopsych.2017.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alexandre Bonnin
- Zilkha Neurogenetic Institute, Department of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|