101
|
Peyvandi S, Baer RJ, Chambers CD, Norton ME, Rajagopal S, Ryckman KK, Moon-Grady A, Jelliffe-Pawlowski LL, Steurer MA. Environmental and Socioeconomic Factors Influence the Live-Born Incidence of Congenital Heart Disease: A Population-Based Study in California. J Am Heart Assoc 2020; 9:e015255. [PMID: 32306820 PMCID: PMC7428546 DOI: 10.1161/jaha.119.015255] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The development of congenital heart disease (CHD) is multifactorial with genetic and environmental influences. We sought to determine the relationship between socioeconomic and environmental factors with the incidence of CHD among live‐born infants in California and to determine whether maternal comorbidities are in the causal pathway. METHODS AND RESULTS This was a population‐based cohort study in California (2007–2012). The primary outcome was having significant CHD. Predictors included socioeconomic status and environmental exposure to pollutants determined by U.S. Census data. A social deprivation index and environmental exposure index was assigned based on neighborhood socioeconomic variables, categorized into 4 quartiles. Quartile 1 was the best with the least exposure to pollutants and social deprivation, and quartile 4 was the worst. Multivariate logistic regression and mediation analyses were performed. Among 2 419 651 live‐born infants, the incidence of CHD was 3.2 per 1000 live births. The incidence of CHD was significantly higher among those in quartile 4 compared with quartile 1 (social deprivation index: 0.35% versus 0.29%; odds ratio [OR], 1.31; 95% CI, 1.21–1.41; environmental exposure index: 0.35% versus 0.29%; OR, 1.23; 95% CI, 1.15–1.31) after adjusting for maternal race/ethnicity and age and accounting for the relationship between the 2 primary predictors. Maternal comorbidities explained 13% (95% CI, 10%–20%) of the relationship between social deprivation index and environmental exposure index with the incidence of CHD. CONCLUSIONS Increased social deprivation and exposure to environmental pollutants are associated with the incidence of live‐born CHD in California. Maternal comorbidities explain some, but not all, of this relationship. These findings identify targets for social policy initiatives to minimize health disparities.
Collapse
Affiliation(s)
- Shabnam Peyvandi
- Division of Cardiology Department of Pediatrics University of California San Francisco Benioff Children's Hospital San Francisco CA.,Department of Epidemiology and Biostatistics University of California San Francisco Benioff Children's Hospital San Francisco CA
| | - Rebecca J Baer
- Obstetrics, Gynecology and Reproductive Sciences University of California San Francisco Benioff Children's Hospital San Francisco CA.,California Preterm Birth Initiative University of California San Francisco Benioff Children's Hospital San Francisco CA.,Department of Pediatrics University of California San Diego La Jolla CA
| | | | - Mary E Norton
- Obstetrics, Gynecology and Reproductive Sciences University of California San Francisco Benioff Children's Hospital San Francisco CA
| | - Satish Rajagopal
- Division of Critical Care University of California San Francisco Benioff Children's Hospital San Francisco CA
| | - Kelli K Ryckman
- Department of Epidemiology College of Public Health University of Iowa Iowa City IA
| | - Anita Moon-Grady
- Division of Cardiology Department of Pediatrics University of California San Francisco Benioff Children's Hospital San Francisco CA
| | - Laura L Jelliffe-Pawlowski
- Department of Epidemiology and Biostatistics University of California San Francisco Benioff Children's Hospital San Francisco CA.,California Preterm Birth Initiative University of California San Francisco Benioff Children's Hospital San Francisco CA
| | - Martina A Steurer
- Division of Critical Care University of California San Francisco Benioff Children's Hospital San Francisco CA.,Department of Epidemiology and Biostatistics University of California San Francisco Benioff Children's Hospital San Francisco CA.,California Preterm Birth Initiative University of California San Francisco Benioff Children's Hospital San Francisco CA
| |
Collapse
|
102
|
Khambadkone SG, Cordner ZA, Tamashiro KLK. Maternal stressors and the developmental origins of neuropsychiatric risk. Front Neuroendocrinol 2020; 57:100834. [PMID: 32084515 PMCID: PMC7243665 DOI: 10.1016/j.yfrne.2020.100834] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.
Collapse
Affiliation(s)
- Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
103
|
Peñalver Bernabé B, Maki PM, Dowty SM, Salas M, Cralle L, Shah Z, Gilbert JA. Precision medicine in perinatal depression in light of the human microbiome. Psychopharmacology (Berl) 2020; 237:915-941. [PMID: 32065252 DOI: 10.1007/s00213-019-05436-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
Perinatal depression is the most common complication of pregnancy and affects the mother, fetus, and infant. Recent preclinical studies and a limited number of clinical studies have suggested an influence of the gut microbiome on the onset and course of mental health disorders. In this review, we examine the current state of knowledge regarding genetics, epigenetics, heritability, and neuro-immuno-endocrine systems biology in perinatal mood disorders, with a particular focus on the interaction between these factors and the gut microbiome, which is mediated via the gut-brain axis. We also provide an overview of experimental and analytical methods that are currently available to researchers interested in elucidating the influence of the gut microbiome on mental health disorders during pregnancy and postpartum.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States.
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shannon M Dowty
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Mariana Salas
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Lauren Cralle
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Zainab Shah
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Scripts Oceanographic Institute, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
104
|
Toso K, de Cock P, Leavey G. Maternal exposure to violence and offspring neurodevelopment: A systematic review. Paediatr Perinat Epidemiol 2020; 34:190-203. [PMID: 32026500 DOI: 10.1111/ppe.12651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/19/2019] [Accepted: 12/25/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Stress during pregnancy is known to affect fetal neurodevelopment. It seems likely therefore that intimate partner violence (IPV) and domestic violence (DV) as extreme stressors will have a similarly adverse effect. OBJECTIVES A systematic review was conducted to assess the association between prenatal exposure to violence for mothers and developmental difficulties in their children. DATA SOURCES PubMed, PsycInfo, CINAHL, ERIC, Science Direct, SCOPUS, PsyARTICLES, Networked Digital Library of Theses and Dissertations, Women's Studies International and Gender Studies Database were all searched using the agreed search terms. STUDY SELECTION AND DATA EXTRACTION We include studies of women who have experienced any violence, fear of violence or aggression while pregnant, including emotional, psychological, physical or sexual violence in the context of IPV or DV. Studies were excluded if the neurodevelopmental outcomes of the offspring were not assessed. Studies from all countries were included, in English or translated to English, and search dates were not restricted. We included all years from inception of the database until the search date. SYNTHESIS Study design and biases, assessment tools, management of confounding, results and overall quality were assessed. RESULTS We identified 11 papers reporting on observational studies. Almost three quarters of the studies found a relationship between prenatal exposure to violence and developmental difficulties in the offspring. Differing assessment tools were used with a range of data collected and not all adjusted their findings for the same confounders. CONCLUSIONS Current evidence on the relationship between prenatal violence exposure, as IPV or DV, and consequent child developmental disorders remains limited. Future research using comprehensive study designs, larger samples and longitudinal follow-up of the offspring could clarify this association. While maternal trauma resulting from exposure to violence may play an important role in childhood development disorders, additional intervening factors on the pathway need further explored.
Collapse
Affiliation(s)
- Kristin Toso
- Bamford Centre for Mental Health and Wellbeing, Ulster University, Coleraine, UK
| | - Paul de Cock
- Bamford Centre for Mental Health and Wellbeing, Ulster University, Coleraine, UK
| | - Gerard Leavey
- Bamford Centre for Mental Health and Wellbeing, Ulster University, Coleraine, UK
| |
Collapse
|
105
|
Howell KR, Law AJ. Neurodevelopmental concepts of schizophrenia in the genome-wide association era: AKT/mTOR signaling as a pathological mediator of genetic and environmental programming during development. Schizophr Res 2020; 217:95-104. [PMID: 31522868 PMCID: PMC7065975 DOI: 10.1016/j.schres.2019.08.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/28/2019] [Accepted: 08/31/2019] [Indexed: 12/14/2022]
Abstract
Normative brain development is contingent on the complex interplay between genes and environment. Schizophrenia (SCZ) is considered a highly polygenic, neurodevelopmental disorder associated with impaired neural circuit development, neurocognitive function and variations in neurotransmitter signaling systems, including dopamine. Significant evidence, accumulated over the last 30 years indicates a role for the in utero environment in SCZ pathophysiology. Emerging data suggests that changes in placental programming and function may mediate the link between genetic risk, early life complications (ELC) and adverse neurodevelopmental outcomes, with risk highlighted in key developmental drivers that converge on AKT/mTOR signaling. In this article we overview select risk genes identified through recent genome-wide association studies of SCZ including AKT3, miR-137, DRD2, and AKT1 itself. We propose that through convergence on AKT/mTOR signaling, these genes are critical factors directing both placentation and neurodevelopment, influencing risk for SCZ through dysregulation of placental function, metabolism and early brain development. We discuss association of risk genes in the context of their known roles in neurodevelopment, placental expression and their possible mechanistic links to SCZ in the broad context of the 'developmental origins of adult disease' construct. Understanding how common genetic variation impacts early fetal programming may advance our knowledge of disease etiology and identify early critical developmental windows for prevention and intervention.
Collapse
Affiliation(s)
| | - Amanda J. Law
- Corresponding Author: Amanda J. Law, PhD, Professor of Psychiatry, Medicine and Cell and Developmental Biology, Nancy L. Gary Endowed Chair in Children’s Mental Disorders Research, University of Colorado, School of Medicine, , Phone: 303-724-4418, Fax: 303-724-4425, 12700 E. 19th Ave., MS 8619, Aurora, CO 80045
| |
Collapse
|
106
|
Zhang W, Ham J, Li Q, Deyssenroth MA, Lambertini L, Huang Y, Tsuchiya KJ, Chen J, Nomura Y. Moderate prenatal stress may buffer the impact of Superstorm Sandy on placental genes: Stress in Pregnancy (SIP) Study. PLoS One 2020; 15:e0226605. [PMID: 31995614 PMCID: PMC6988921 DOI: 10.1371/journal.pone.0226605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 11/29/2019] [Indexed: 12/26/2022] Open
Abstract
The placenta plays a central role in the epigenetic programming of neurodevelopment by prenatal stress (PS), but this pathway is not fully understood. It difficult to study in humans because the conditions for intense, traumatic PS are almost impossible to create ethically. This study was able to capitalize on a 2012 disaster that hit New York, Superstorm Sandy, to examine the impact of traumatic stress on placental gene expression while also examining normative PS, and compare the two. Of the 303 expectant mothers participating in the Stress in Pregnancy Study, 95 women were pregnant when Superstorm Sandy struck. During their pregnancy, participants completed self-report measures of PS and distress that were combined, using latent profile analysis, into one global indicator of normative PS. Placental tissue was collected at delivery and frozen for storage. RNA expression was assessed for 40 placental genes known to associate with the stress response system and neurodevelopment in offspring. Results showed that normative PS increased expression of just MECP2, HSD11B2, and ZNF507, whereas Superstorm Sandy PS decreased expression of CDKL5, CFL1, DYRK1A, HSD11B2, MAOA, MAOB, NCOR1, and ZNF507. Interaction analyses indicated that Superstorm Sandy PS was associated with decreased gene expression for the low and high PS group for CFL1, DYRK1A, HSD11B2, MAOA, and NCOR1 and increased expression for the moderate PS group for FOXP1, NR3C1, and NR3C2. This study supports the idea that a moderate amount of normative PS may buffer the impact of traumatic PS, in this case caused by Superstorm Sandy, on placental gene expression, which suggests that the placenta itself mirrors the organism's ability to develop an epigenetic resilience to, and inoculation from, stress.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Psychology, Queens College, CUNY, New York, NY, United States of America
- Department of Psychology, New Jersey City University, Jersey City, NJ, United States of America
| | - Jacob Ham
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Qian Li
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Maya A. Deyssenroth
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Luca Lambertini
- Department of Medicine, Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Yonglin Huang
- Department of Psychology, Queens College, CUNY, New York, NY, United States of America
- Department of Psychology, The Graduate Center, CUNY, New York, NY, United States of America
| | - Kenji J. Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Yoko Nomura
- Department of Psychology, Queens College, CUNY, New York, NY, United States of America
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Psychology, The Graduate Center, CUNY, New York, NY, United States of America
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
107
|
Arnegard ME, Whitten LA, Hunter C, Clayton JA. Sex as a Biological Variable: A 5-Year Progress Report and Call to Action. J Womens Health (Larchmt) 2020; 29:858-864. [PMID: 31971851 DOI: 10.1089/jwh.2019.8247] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A little over 5 years ago, the U.S. National Institutes of Health (NIH) announced the intention to develop policies to require applicants to report plans to balance male and female cells and animals in preclinical investigations. Soon thereafter, the NIH issued a request for information from the scientific community and consulted with various stakeholders. The feedback received was considered during development of policy requiring the consideration of sex as a biological variable (SABV) in NIH-funded research on vertebrate animals and humans, which went into effect for applications due on or after January 25, 2016. We identified NIH programs related to SABV and reviewed SABV-relevant scientific literature. We find that the application of SABV throughout the research process can serve as a guiding principle to improve the value of biomedical science. The NIH is engaged in ongoing efforts to develop resources to help investigators consider SABV in their research. We also provide an update on lessons learned, highlight ways that different disciplines consider SABV, and describe the opportunities for scientific discovery that applying SABV offers. We call on NIH's various stakeholders to redouble their efforts to integrate SABV throughout the biomedical research enterprise. Sex- and gender-aware investigations are critical to the conduct of rigorous and transparent science and the advancement of personalized medicine. This kind of research achieves its greatest potential when sex and gender considerations are integrated into the biomedical research enterprise in an end-to-end manner, from basic and preclinical investigations, through translational and clinical research, to improved health care delivery.
Collapse
Affiliation(s)
- Matthew E Arnegard
- Office of Research on Women's Health, National Institutes of Health, Bethesda, Maryland
| | | | - Chyren Hunter
- Office of Research on Women's Health, National Institutes of Health, Bethesda, Maryland
| | - Janine Austin Clayton
- Office of Research on Women's Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
108
|
Maternal inflammation has a profound effect on cortical interneuron development in a stage and subtype-specific manner. Mol Psychiatry 2020; 25:2313-2329. [PMID: 31595033 PMCID: PMC7515848 DOI: 10.1038/s41380-019-0539-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/10/2019] [Accepted: 09/24/2019] [Indexed: 01/21/2023]
Abstract
Severe infections during pregnancy are one of the major risk factors for cognitive impairment in the offspring. It has been suggested that maternal inflammation leads to dysfunction of cortical GABAergic interneurons that in turn underlies cognitive impairment of the affected offspring. However, the evidence comes largely from studies of adult or mature brains and how the impairment of inhibitory circuits arises upon maternal inflammation is unknown. Here we show that maternal inflammation affects multiple steps of cortical GABAergic interneuron development, i.e., proliferation of precursor cells, migration and positioning of neuroblasts, as well as neuronal maturation. Importantly, the development of distinct subtypes of cortical GABAergic interneurons was discretely impaired as a result of maternal inflammation. This translated into a reduction in cell numbers, redistribution across cortical regions and layers, and changes in morphology and cellular properties. Furthermore, selective vulnerability of GABAergic interneuron subtypes was associated with the stage of brain development. Thus, we propose that maternally derived insults have developmental stage-dependent effects, which contribute to the complex etiology of cognitive impairment in the affected offspring.
Collapse
|
109
|
Walsh K, McCormack CA, Webster R, Pinto A, Lee S, Feng T, Krakovsky HS, O'Grady SM, Tycko B, Champagne FA, Werner EA, Liu G, Monk C. Maternal prenatal stress phenotypes associate with fetal neurodevelopment and birth outcomes. Proc Natl Acad Sci U S A 2019; 116:23996-24005. [PMID: 31611411 PMCID: PMC6883837 DOI: 10.1073/pnas.1905890116] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Maternal prenatal stress influences offspring neurodevelopment and birth outcomes including the ratio of males to females born; however, there is limited understanding of what types of stress matter, and for whom. Using a data-driven approach with 27 variables from questionnaires, ambulatory diaries, and physical assessments collected early in the singleton pregnancies of 187 women, 3 latent profiles of maternal prenatal stress emerged that were differentially associated with sex at birth, birth outcomes, and fetal neurodevelopment. Most women (66.8%) were in the healthy group (HG); 17.1% were in the psychologically stressed group (PSYG), evidencing clinically meaningful elevations in perceived stress, depression, and anxiety; and 16% were in the physically stressed group (PHSG) with relatively higher ambulatory blood pressure and increased caloric intake. The population normative male:female secondary sex ratio (105:100) was lower in the PSYG (2:3) and PHSG (4:9), and higher in the HG (23:18), consistent with research showing diminished male births in maternal stress contexts. PHSG versus HG infants were born 1.5 wk earlier (P < 0.05) with 22% compared to 5% born preterm. PHSG versus HG fetuses had decreased fetal heart rate-movement coupling (P < 0.05), which may indicate slower central nervous system development, and PSYG versus PHSG fetuses had more birth complications, consistent with previous findings among offspring of women with psychiatric illness. Social support most strongly differentiated the HG, PSYG, and PHSG groups, and higher social support was associated with increased odds of male versus female births. Stress phenotypes in pregnant women are associated with male vulnerability and poor fetal outcomes.
Collapse
Affiliation(s)
- Kate Walsh
- Ferkauf Graduate School of Psychology, Yeshiva University, The Bronx, NY 10461
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Clare A McCormack
- Center for Science and Society, Columbia University, New York, NY 10027
| | - Rachel Webster
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032
| | - Anita Pinto
- Data Science, Columbia University, New York, NY 10027
| | - Seonjoo Lee
- Division of Behavioral Medicine, New York State Psychiatric Institute, New York, NY 10032
- Department of Biostatistics (in Psychiatry), Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Tianshu Feng
- Department of Biostatistics (in Psychiatry), Mailman School of Public Health, Columbia University, New York, NY 10032
| | - H Sloan Krakovsky
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032
| | - Sinclaire M O'Grady
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032
| | - Benjamin Tycko
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, NJ 07110
| | - Frances A Champagne
- Department of Psychiatry, Columbia University, New York, NY 10032
- Department of Psychology, University of Texas at Austin, Austin, TX 78712
| | - Elizabeth A Werner
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Grace Liu
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Catherine Monk
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032;
- Division of Behavioral Medicine, New York State Psychiatric Institute, New York, NY 10032
- Department of Psychiatry, Columbia University, New York, NY 10032
| |
Collapse
|
110
|
Lipner E, Murphy SK, Ellman LM. Prenatal Maternal Stress and the Cascade of Risk to Schizophrenia Spectrum Disorders in Offspring. Curr Psychiatry Rep 2019; 21:99. [PMID: 31522269 PMCID: PMC7043262 DOI: 10.1007/s11920-019-1085-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Disruptions in fetal development (via genetic and environmental pathways) have been consistently associated with risk for schizophrenia in a variety of studies. Although multiple obstetric complications (OCs) have been linked to schizophrenia, this review will discuss emerging evidence supporting the role of prenatal maternal stress (PNMS) in the etiology of schizophrenia spectrum disorders (SSD). In addition, findings linking PNMS to intermediate phenotypes of the disorder, such as OCs and premorbid cognitive, behavioral, and motor deficits, will be reviewed. Maternal immune and endocrine dysregulation will also be explored as potential mechanisms by which PNMS confers risk for SSD. RECENT FINDINGS PNMS has been linked to offspring SSD; however, findings are mixed due to inconsistent and retrospective assessments of PNMS and lack of specificity about SSD outcomes. PNMS is also associated with various intermediate phenotypes of SSD (e.g., prenatal infection/inflammation, decreased fetal growth, hypoxia-related OCs). Recent studies continue to elucidate the impact of PNMS while considering the moderating roles of fetal sex and stress timing, but it is still unclear which aspects of PNMS (e.g., type, timing) confer risk for SSD specifically. PNMS increases risk for SSD, but only in a small portion of fetuses exposed to PNMS. Fetal sex, genetics, and other environmental factors, as well as additional pre- and postnatal insults, likely contribute to the PNMS-SSD association. Longitudinal birth cohort studies are needed to prospectively illuminate the mechanisms that account for the variability in outcomes following PNMS.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA
| | - Shannon K Murphy
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA.
| |
Collapse
|
111
|
Knöfler M, Haider S, Saleh L, Pollheimer J, Gamage TKJB, James J. Human placenta and trophoblast development: key molecular mechanisms and model systems. Cell Mol Life Sci 2019; 76:3479-3496. [PMID: 31049600 PMCID: PMC6697717 DOI: 10.1007/s00018-019-03104-6] [Citation(s) in RCA: 376] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/16/2022]
Abstract
Abnormal placentation is considered as an underlying cause of various pregnancy complications such as miscarriage, preeclampsia and intrauterine growth restriction, the latter increasing the risk for the development of severe disorders in later life such as cardiovascular disease and type 2 diabetes. Despite their importance, the molecular mechanisms governing human placental formation and trophoblast cell lineage specification and differentiation have been poorly unravelled, mostly due to the lack of appropriate cellular model systems. However, over the past few years major progress has been made by establishing self-renewing human trophoblast stem cells and 3-dimensional organoids from human blastocysts and early placental tissues opening the path for detailed molecular investigations. Herein, we summarize the present knowledge about human placental development, its stem cells, progenitors and differentiated cell types in the trophoblast epithelium and the villous core. Anatomy of the early placenta, current model systems, and critical key regulatory factors and signalling cascades governing placentation will be elucidated. In this context, we will discuss the role of the developmental pathways Wingless and Notch, controlling trophoblast stemness/differentiation and formation of invasive trophoblast progenitors, respectively.
Collapse
Affiliation(s)
- Martin Knöfler
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria.
| | - Sandra Haider
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Leila Saleh
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Jürgen Pollheimer
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Teena K J B Gamage
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
112
|
Placental programming of neuropsychiatric disease. Pediatr Res 2019; 86:157-164. [PMID: 31003234 DOI: 10.1038/s41390-019-0405-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [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
|
113
|
Grandjean P, Abdennebi-Najar L, Barouki R, Cranor CF, Etzel RA, Gee D, Heindel JJ, Hougaard KS, Hunt P, Nawrot TS, Prins GS, Ritz B, Soffritti M, Sunyer J, Weihe P. Timescales of developmental toxicity impacting on research and needs for intervention. Basic Clin Pharmacol Toxicol 2019; 125 Suppl 3:70-80. [PMID: 30387920 PMCID: PMC6497561 DOI: 10.1111/bcpt.13162] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/29/2018] [Indexed: 12/28/2022]
Abstract
Much progress has happened in understanding developmental vulnerability to preventable environmental hazards. Along with the improved insight, the perspective has widened, and developmental toxicity now involves latent effects that can result in delayed adverse effects in adults or at old age and additional effects that can be transgenerationally transferred to future generations. Although epidemiology and toxicology to an increasing degree are exploring the adverse effects from developmental exposures in human beings, the improved documentation has resulted in little progress in protection, and few environmental chemicals are currently regulated to protect against developmental toxicity, whether it be neurotoxicity, endocrine disruption or other adverse outcome. The desire to obtain a high degree of certainty and verification of the evidence used for decision-making must be weighed against the costs and necessary duration of research, as well as the long-term costs to human health because of delayed protection of vulnerable early-life stages of human development and, possibly, future generations. Although two-generation toxicology tests may be useful for initial test purposes, other rapidly emerging tools need to be seriously considered from computational chemistry and metabolomics to CLARITY-BPA-type designs, big data and population record linkage approaches that will allow efficient generation of new insight; epigenetic mechanisms may necessitate a set of additional regulatory tests to reveal such effects. As reflected by the Prenatal Programming and Toxicity (PPTOX) VI conference, the current scientific understanding and the timescales involved require an intensified approach to protect against preventable adverse health effects that can harm the next generation and generations to come. While further research is needed, the main emphasis should be on research translation and timely public health intervention to avoid serious, irreversible and perhaps transgenerational harm.
Collapse
Affiliation(s)
- Philippe Grandjean
- Department of Environmental Medicine, University of Southern Denmark, Odense, Denmark
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Robert Barouki
- INSERM UMR-S 1124, Université Paris Descartes, Paris, France
| | - Carl F Cranor
- Department of Philosophy, University of California, Riverside, California
| | - Ruth A Etzel
- Milken Institute, School of Public Health, The George Washington University, Washington, District of Columbia
| | - David Gee
- Institute of Environment, Health and Societies, Brunel University, London, UK
| | - Jerrold J Heindel
- Program in Endocrine Disruption Strategies, Commonweal, Bolinas, California
| | - Karin S Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Patricia Hunt
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Department of Public Health, Leuven University, Leuven, Belgium
| | - Gail S Prins
- Chicago Center for Health and Environment (CACHET), University of Illinois at Chicago, Chicago, Illinois
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California
| | - Morando Soffritti
- Ramazzini Institute, Bologna, Italy
- European Foundation for Cancer Research, Environmental and Occupational Diseases "Ruberti Schileo", Treviso, Italy
| | - Jordi Sunyer
- ISGlobal, Centre for Research in Environmental Epidemiology, Barcelona, Catalonia, Spain
| | - Pal Weihe
- Department of Public Health and Occupational Medicine, Tórshavn, Faroe Islands
| |
Collapse
|
114
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
115
|
Microglia along sex lines: From brain colonization, maturation and function, to implication in neurodevelopmental disorders. Semin Cell Dev Biol 2019; 94:152-163. [PMID: 31201858 DOI: 10.1016/j.semcdb.2019.06.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
In addition to their traditional role as immune sentinels, recent discoveries over the last decade have shown that microglial functions now include regulation of neuronal/glial cell migration, differentiation and maturation, as well as neuronal network formation. It was thus proposed that disruption of these microglial roles, during critical periods of brain development, could lead to the pathological onset of several neurodevelopmental disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, epilepsy, schizophrenia, and major depressive disorder. The prevalence of these disorders exhibits a clear distinction along sex lines with very little known about the mechanisms underlying this difference. One of the fundamental discoveries that arose from recent research into the physiological roles of microglia in neurodevelopment is their sexual dimorphism, raising the intriguing possibility that sex differences in microglial colonization, maturation and/or function in the developing brain could underlie the emergence of various neurodevelopmental disorders. This review discusses the physiological roles of microglia across neurodevelopment, these roles in the two sexes, and the recent evidence that microglial sexually dimorphic nature may contribute, at least partially, to neurodevelopmental disorders.
Collapse
|
116
|
Integrated analysis of environmental and genetic influences on cord blood DNA methylation in new-borns. Nat Commun 2019; 10:2548. [PMID: 31186427 PMCID: PMC6559955 DOI: 10.1038/s41467-019-10461-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Epigenetic processes, including DNA methylation (DNAm), are among the mechanisms allowing integration of genetic and environmental factors to shape cellular function. While many studies have investigated either environmental or genetic contributions to DNAm, few have assessed their integrated effects. Here we examine the relative contributions of prenatal environmental factors and genotype on DNA methylation in neonatal blood at variably methylated regions (VMRs) in 4 independent cohorts (overall n = 2365). We use Akaike’s information criterion to test which factors best explain variability of methylation in the cohort-specific VMRs: several prenatal environmental factors (E), genotypes in cis (G), or their additive (G + E) or interaction (GxE) effects. Genetic and environmental factors in combination best explain DNAm at the majority of VMRs. The CpGs best explained by either G, G + E or GxE are functionally distinct. The enrichment of genetic variants from GxE models in GWAS for complex disorders supports their importance for disease risk. Environmental influences during prenatal development may have implications for health and disease later in life. Here, Czamara et al. assess DNA methylation in cord blood from new-born under various models including environmental and genetic effects individually and their additive or interaction effects.
Collapse
|
117
|
Chen HJ, Gur TL. Intrauterine Microbiota: Missing, or the Missing Link? Trends Neurosci 2019; 42:402-413. [PMID: 31053242 PMCID: PMC6604064 DOI: 10.1016/j.tins.2019.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 12/29/2022]
Abstract
The intrauterine environment provides a key interface between the mother and the developing fetus during pregnancy, and is a target for investigating mechanisms of fetal programming. Studies have demonstrated an association between prenatal stress and neurodevelopmental disorders. The role of the intrauterine environment in mediating this effect is still being elucidated. In this review, we discuss emerging preclinical and clinical evidence suggesting the existence of microbial communities in utero. We also outline possible mechanisms of bacterial translocation to the intrauterine environment and immune responses to the presence of microbes or microbial components. Lastly, we overview the effects of intrauterine inflammation on neurodevelopment. We hypothesize that maternal gestational stress leads to disruptions in the maternal oral, gut, and vaginal microbiome that may lead to the translocation of bacteria to the intrauterine environment, eliciting an inflammatory response and resulting in deficits in neurodevelopment.
Collapse
Affiliation(s)
- Helen J Chen
- Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - Tamar L Gur
- Department of Psychiatry and Behavioral Health, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Obstetrics and Gynecology, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Institute of Behavioral Medicine Research, Wexner Medical Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
118
|
Abstract
PURPOSE OF REVIEW With the incidence of neurodevelopmental disorders on the rise, it is imperative to identify and understand the mechanisms by which environmental contaminants can impact the developing brain and heighten risk. Here, we report on recent findings regarding novel mechanisms of developmental neurotoxicity and highlight chemicals of concern, beyond traditionally defined neurotoxicants. RECENT FINDINGS The perinatal window represents a critical and extremely vulnerable period of time during which chemical insult can alter the morphological and functional trajectory of the developing brain. Numerous chemical classes have been associated with alterations in neurodevelopment including metals, solvents, pesticides, and, more recently, endocrine-disrupting compounds. Although mechanisms of neurotoxicity have traditionally been identified as pathways leading to neuronal cell death, neuropathology, or severe neural injury, recent research highlights alternative mechanisms that result in more subtle but consequential changes in the brain and behavior. These emerging areas of interest include neuroendocrine and immune disruption, as well as indirect toxicity via actions on other organs such as the gut and placenta. Understanding of the myriad ways in which the developing brain is vulnerable to chemical exposures has grown tremendously over the past decade. Further progress and implementation in risk assessment is critical to reducing risk of neurodevelopmental disorders.
Collapse
|
119
|
Morales-Rubio RA, Alvarado-Cruz I, Manzano-León N, Andrade-Oliva MDLA, Uribe-Ramirez M, Quintanilla-Vega B, Osornio-Vargas Á, De Vizcaya-Ruiz A. In utero exposure to ultrafine particles promotes placental stress-induced programming of renin-angiotensin system-related elements in the offspring results in altered blood pressure in adult mice. Part Fibre Toxicol 2019; 16:7. [PMID: 30691489 PMCID: PMC6350404 DOI: 10.1186/s12989-019-0289-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/13/2019] [Indexed: 12/16/2022] Open
Abstract
Background Exposure to particulate matter (PM) is associated with an adverse intrauterine environment, which can promote adult cardiovascular disease (CVD) risk. Ultrafine particles (UFP) (small size and large surface area/mass ratio) are systemically distributed, induce inflammation and oxidative stress, and have been associated with vascular endothelial dysfunction and arterial vasoconstriction, increasing hypertension risk. Placental stress and alterations in methylation of promoter regions of renin-angiotensin system (RAS)-related elements could be involved in UFP exposure-related programming of hypertension. We investigated whether in utero UFP exposure promotes placental stress by inflammation and oxidative stress, alterations in hydroxysteroid dehydrogenase 11b-type 2 (HSD11B2) and programming of RAS-related elements, and result in altered blood pressure in adult offspring. UFP were collected from ambient air using an aerosol concentrator and physicochemically characterized. Pregnant C57BL/6J pun/pun female mice were exposed to collected UFP (400 μg/kg accumulated dose) by intratracheal instillation and compared to control (nonexposed) and sterile H2O (vehicle) exposed mice. Embryo reabsorption and placental stress by measurement of the uterus, placental and fetal weights, dam serum and fetal cortisol, placental HSD11B2 DNA methylation and protein levels, were evaluated. Polycyclic aromatic hydrocarbon (PAH) biotransformation (CYP1A1 and NQO1 (NAD(P)H dehydrogenase (quinone)1)) enzymes, inflammation and oxidative stress in placentas and fetuses were measured. Postnatal day (PND) 50 in male offspring blood pressure was measured. Methylation and protein expression of (RAS)-related elements, angiotensin II receptor type 1 (AT1R) and angiotensin I-converting enzyme (ACE) in fetuses and lungs of PND 50 male offspring were also assessed. Results In utero UFP exposure induced placental stress as indicated by an increase in embryo reabsorption, decreases in the uterus, placental, and fetal weights, and HSD11B2 hypermethylation and protein downregulation. In utero UFP exposure induced increases in the PAH-biotransforming enzymes, intrauterine oxidative damage and inflammation and stimulated programming and activation of AT1R and ACE, which resulted in increased blood pressure in the PND 50 male offspring. Conclusions In utero UFP exposure promotes placental stress through inflammation and oxidative stress, and programs RAS-related elements that result in altered blood pressure in the offspring. Exposure to UFP during fetal development could influence susceptibility to CVD in adulthood. Electronic supplementary material The online version of this article (10.1186/s12989-019-0289-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Russell A Morales-Rubio
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Isabel Alvarado-Cruz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Natalia Manzano-León
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Maria-de-Los-Angeles Andrade-Oliva
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Marisela Uribe-Ramirez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Betzabet Quintanilla-Vega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | | | - Andrea De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México.
| |
Collapse
|
120
|
Edlow AG, Glass RM, Smith CJ, Tran PK, James K, Bilbo S. Placental Macrophages: A Window Into Fetal Microglial Function in Maternal Obesity. Int J Dev Neurosci 2018; 77:60-68. [PMID: 30465871 DOI: 10.1016/j.ijdevneu.2018.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 01/18/2023] Open
Abstract
Fetal placental macrophages and microglia (resident brain macrophages) have a common origin in the fetal yolk sac. Yolk-sac-derived macrophages comprise the permanent pool of brain microglia throughout an individual's lifetime. Inappropriate fetal microglial priming may therefore have lifelong neurodevelopmental consequences, but direct evaluation of microglial function in a living fetus or neonate is impossible. We sought to test the hypothesis that maternal obesity would prime both placental macrophages and fetal brain microglia to overrespond to an immune challenge, thus providing a window into microglial function using placental cells. Obesity was induced in C57BL/6 J mice using a 60% high-fat diet. On embryonic day 17.5, fetal brain microglia and corresponding CD11b + placental cells were isolated from fresh tissue. Cells were treated with media or lipopolysaccharide (LPS). Tumor necrosis factor-alpha (TNF-α) production by stimulated and unstimulated cells was quantified via ELISA. We demonstrate for the first time that the proinflammatory cytokine production of CD11b + placental cells is strongly correlated with that of brain microglia (Spearman's ρ = 0.73, p = 0.002) in the setting of maternal obesity. Maternal obesity-exposed CD11b + cells had an exaggerated response to LPS compared to controls, with a 5.1-fold increase in TNF-α production in placentas (p = 0.003) and 3.8-fold increase in TNF-α production in brains (p = 0.002). In sex-stratified analyses, only male obesity-exposed brains and placentas had significant increase in TNF-α production in response to LPS. Taken together, these data suggest that maternal obesity primes both placental macrophages and fetal brain microglia to overproduce a proinflammatory cytokine in response to immune challenge. Male brain and placental immune response is more marked than female in this setting. Given that fetal microglial priming may impact neuroimmune function throughout the lifespan, these data could provide insight into the male predominance of certain neurodevelopmental morbidities linked to maternal obesity, including cognitive dysfunction, autism spectrum disorder, and ADHD. Placental CD11b+ macrophages may have the potential to serve as an accessible biomarker of aberrant fetal brain immune activation in maternal obesity. This finding may have broader implications for assaying the impact of other maternal exposures on fetal brain development.
Collapse
Affiliation(s)
- Andrea G Edlow
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Ruthy M Glass
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Caroline J Smith
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Phuong Kim Tran
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Kaitlyn James
- Massachusetts General Hospital, Deborah Kelly Center for Outcomes Research
| | - Staci Bilbo
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| |
Collapse
|
121
|
Schroeder M, Jakovcevski M, Polacheck T, Drori Y, Ben-Dor S, Röh S, Chen A. Sex dependent impact of gestational stress on predisposition to eating disorders and metabolic disease. Mol Metab 2018; 17:1-16. [PMID: 30174229 PMCID: PMC6197785 DOI: 10.1016/j.molmet.2018.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Vulnerability to eating disorders (EDs) is broadly assumed to be associated with early life stress. However, a careful examination of the literature shows that susceptibility to EDs may depend on the type, severity and timing of the stressor and the sex of the individual. We aimed at exploring the link between chronic prenatal stress and predisposition to EDs and metabolic disease. METHODS We used a chronic variable stress protocol during gestation to explore the metabolic response of male and female offspring to food restriction (FR), activity-based anorexia (ABA), binge eating (BE) and exposure to high fat (HF) diet. RESULTS Contrary to controls, prenatally stressed (PNS) female offspring showed resistance to ABA and BE and displayed a lower metabolic rate leading to hyperadiposity and obesity on HF diet. Male PNS offspring showed healthy responses to FR and ABA, increased propensity to binge and improved coping with HF compared to controls. We found that long-lasting abnormal responses to metabolic challenge are linked to fetal programming and adult hypothalamic dysregulation in PNS females, resulting from sexually dimorphic adaptations in placental methylation and gene expression. CONCLUSIONS Our results show that maternal stress may have variable and even opposing effects on ED risk, depending on the ED and the sex of the offspring.
Collapse
Affiliation(s)
- Mariana Schroeder
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Tamar Polacheck
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Yonat Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| |
Collapse
|
122
|
Zhang J, Luo W, Huang P, Peng L, Huang Q. Maternal C-reactive protein and cytokine levels during pregnancy and the risk of selected neuropsychiatric disorders in offspring: A systematic review and meta-analysis. J Psychiatr Res 2018; 105:86-94. [PMID: 30212728 DOI: 10.1016/j.jpsychires.2018.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
The pathophysiology of neuropsychiatric disorders is unclear. Increasing evidence has suggested maternal immune system dysregulation may be implicated in offspring's early brain development and related to an increased risk of neuropsychiatric disorders in offspring. The primary objective of this meta-analysis was to investigate the association of maternal CRP and cytokine levels with offspring's neuropsychiatric disorders. We identified relevant studies following a search of PubMed, Web of Science, EMbase database between January 1971 and February 2018. A meta-analysis was performed on studies which reported the association of prenatal maternal peripheral blood concentrations of CRP and cytokines with offspring's neuropsychiatric disorders. Results were reported according to PRISMA statement. Fifteen studies (six for maternal CRP, nine for maternal cytokines) were included in the meta-analysis, of which 80% were of high methodological quality. Random-effect meta-analysis showed that increasing maternal CRP (OR = 1.31, 95% CI 1.11-1.55, SMD = 0.15, 95% CI 0.06-0.24, P < 0.01), pro-inflammatory cytokine interleukin (IL)-8 (OR = 1.64, 95% CI 1.06-2.55, SMD = 0.27, 95% CI 0.03-0.52, P = 0.03) and anti-inflammatory cytokine IL-10 (OR = 2.16, 95% CI 1.30-3.59, SMD = 0.43, 95% CI 0.14-0.71, P < 0.01) were significantly associated with schizophrenia in offspring. The finding of our meta-analysis has identified significantly altered maternal CRP and cytokine concentrations in schizophrenia, strengthening evidence of maternal immune system dysregulation in neuropsychiatric disorders where inflammatory signals dominate.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China; Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Wanjun Luo
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Pengcheng Huang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Linrui Peng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qitao Huang
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China; Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
123
|
Korzeniewski SJ, Slaughter J, Lenski M, Haak P, Paneth N. The complex aetiology of cerebral palsy. Nat Rev Neurol 2018; 14:528-543. [PMID: 30104744 DOI: 10.1038/s41582-018-0043-6] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cerebral palsy (CP) is the most prevalent, severe and costly motor disability of childhood. Consequently, CP is a public health priority for prevention, but its aetiology has proved complex. In this Review, we summarize the evidence for a decline in the birth prevalence of CP in some high-income nations, describe the epidemiological evidence for risk factors, such as preterm delivery and fetal growth restriction, genetics, pregnancy infection and other exposures, and discuss the success achieved so far in prevention through the use of magnesium sulfate in preterm labour and therapeutic hypothermia for birth-asphyxiated infants. We also consider the complexities of disentangling prenatal and perinatal influences, and of establishing subtypes of the disorder, with a view to accelerating the translation of evidence into the development of strategies for the prevention of CP.
Collapse
Affiliation(s)
- Steven J Korzeniewski
- Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Jaime Slaughter
- Department of Health Systems and Sciences Research and Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, PA, USA
| | - Madeleine Lenski
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Peterson Haak
- Michigan Department of Health and Human Services, Lansing, MI, USA
| | - Nigel Paneth
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
124
|
Lodha A, Lakhani J, Ediger K, Tang S, Lodha A, Gandhi V, Creighton D. Do preterm infants with a birth weight ≤1250 g born to single-parent families have poorer neurodevelopmental outcomes at age 3 than those born to two-parent families? J Perinatol 2018; 38:900-907. [PMID: 29740187 DOI: 10.1038/s41372-018-0118-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 03/17/2018] [Accepted: 03/26/2018] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Investigate neurodevelopmental outcomes at 3 years corrected age in infants with a birth weight ≤1250 g born to single parents. STUDY DESIGN Infants born between 1995 and 2010 with a birth weight ≤1250 g were considered eligible. Primary outcome was neurodevelopmental impairment; considered present if a child had any of the following: cerebral palsy, cognitive delay, visual impairment, or deafness/neurosensory hearing impairment. Univariate and multivariate analyses were performed. RESULT A total of 1900 infants were eligible for inclusion. Follow-up data were available for 1395; 88 were born to a single parent. Infants in the single-parent group had higher mortality (18% vs. 11%, p = 0.009), IQ ≥1 SD below the mean (40% vs. 21%, p = 0.001) and any neurodevelopmental impairment (47% vs. 29%, p = 0.003). Single-parent family status, maternal education, bronchopulmonary dysplasia and severe neurological injury were significant predictors of intellectual impairment at 3 years corrected age. CONCLUSION Preterm infants with a birth weight ≤1250 g born to single parents at birth have poorer intellectual functioning at 3 years corrected age.
Collapse
Affiliation(s)
- Abhay Lodha
- Alberta Health Services, University of Calgary, Calgary, AL, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AL, Canada. .,Department of Pediatrics, University of Calgary, Calgary, AL, Canada. .,Department of Community Health Sciences, University of Calgary, Calgary, AL, Canada.
| | - Jahan Lakhani
- Alberta Health Services, University of Calgary, Calgary, AL, Canada
| | - Krystyna Ediger
- Department of Pediatrics, University of Calgary, Calgary, AL, Canada
| | - Selphee Tang
- Alberta Health Services, University of Calgary, Calgary, AL, Canada
| | - Arijit Lodha
- Graduate Department, University of Calgary, Calgary, AL, Canada
| | - Vardhil Gandhi
- Graduate Department, University of Calgary, Calgary, AL, Canada
| | - Dianne Creighton
- Alberta Health Services, University of Calgary, Calgary, AL, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AL, Canada
| |
Collapse
|
125
|
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
|
126
|
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]
|
127
|
Convergence of placenta biology and genetic risk for schizophrenia. Nat Med 2018; 24:792-801. [DOI: 10.1038/s41591-018-0021-y] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/16/2018] [Indexed: 01/16/2023]
|
128
|
Placental miR-340 mediates vulnerability to activity based anorexia in mice. Nat Commun 2018; 9:1596. [PMID: 29686286 PMCID: PMC5913294 DOI: 10.1038/s41467-018-03836-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
Anorexia nervosa (AN) is a devastating eating disorder characterized by self-starvation that mainly affects women. Its etiology is unknown, which impedes successful treatment options leading to a limited chance of full recovery. Here, we show that gestation is a vulnerable window that can influence the predisposition to AN. By screening placental microRNA expression of naive and prenatally stressed (PNS) fetuses and assessing vulnerability to activity-based anorexia (ABA), we identify miR-340 as a sexually dimorphic regulator involved in prenatal programming of ABA. PNS caused gene-body hypermethylation of placental miR-340, which is associated with reduced miR-340 expression and increased protein levels of several target transcripts, GR, Cry2 and H3F3b. MiR-340 is linked to the expression of several nutrient transporters both in mice and human placentas. Using placenta-specific lentiviral transgenes and embryo transfer, we demonstrate the key role miR-340 plays in the mechanism involved in early life programming of ABA. Anorexia nervosa is characterised by self-starvation but its etiology is not completely understood. Here the authors describe how prenatal stress can induce activity-based anorexia in the offspring during early adulthood by upregulating miR-340 expression in the placenta that affects expression of nutrient transporters.
Collapse
|
129
|
Peretti S, Mariano M, Mazzocchetti C, Mazza M, Pino MC, Verrotti Di Pianella A, Valenti M. Diet: the keystone of autism spectrum disorder? Nutr Neurosci 2018; 22:825-839. [PMID: 29669486 DOI: 10.1080/1028415x.2018.1464819] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Children with autism are characterized by an impairment of social interaction and repetitive patterns of behaviour. Autism is a heterogeneous span of disorders with unknown aetiology. Research has grown significantly and has suggested that environmental risk factors acting during the prenatal period could influence the neurodevelopment of offspring. The literature suggests that the maternal diet during pregnancy has a fundamental role in the etiopathogenesis of autism. Indeed, a maternal diet that is high in some nutrients has been associated with an increase or reduction in the risk of develop Autism Spectrum Disorders (ASD). The diet of ASD children is also a key factor for the worsening of ASD symptoms. Children with autism have food selectivity and limited diets due to smell, taste, or other characteristics of foods. This determines eating routines and food intake patterns, with consequent deficiency or excess of some aliments. Several studies have tried to show a possible relationship between nutritional status and autism. In this review we describe, emphasizing the limits and benefits, the main current empirical studies that have examined the role of maternal diet during gestation and diet of ASD children as modifiable risk factors at the base of development or worsening of symptoms of autism.
Collapse
Affiliation(s)
- S Peretti
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - M Mariano
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - C Mazzocchetti
- Department of Pediatrics, San Salvatore Hospital, University of L'Aquila, Località Coppito, L'Aquila 67100, Italy
| | - M Mazza
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - M C Pino
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - A Verrotti Di Pianella
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Department of Pediatrics, San Salvatore Hospital, University of L'Aquila, Località Coppito, L'Aquila 67100, Italy
| | - M Valenti
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| |
Collapse
|
130
|
Pehme PM, Zhang W, Finik J, Pritchett A, Buthmann J, Dana K, Hao K, Nomura Y. Placental MAOA expression mediates prenatal stress effects on temperament in 12-month-olds. INFANT AND CHILD DEVELOPMENT 2018; 27. [PMID: 30505241 DOI: 10.1002/icd.2094] [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] [Indexed: 12/22/2022]
Abstract
The placenta adapts to maternal environment and its alterations may have a lasting impact on child's temperament development. Prenatal stress has been linked to both a downregulation of monoamine oxidase A (MAOA) gene expression in the placenta and to difficult temperament. Capitalizing on an ongoing longitudinal study, we analysed data from 95 mother-child dyads to investigate whether MAOA mediates the association between prenatal stress and infant temperament. Prenatal stress was defined as exposure to Superstorm Sandy (Sandy) during pregnancy. Infant temperament was measured by Infant Behaviour Questionnaire-Revised. MAOA gene expression was quantified in placenta tissue. The Smiling and Laughter subscale score was independently associated with Sandy exposure and MAOA placental gene expression. Mediation analysis confirmed that MAOA expression partially mediated the relationship between Sandy and Smiling and Laughter subscale, suggesting that in utero exposure to Sandy could induce lower frequency of smiling and laughter via downregulation of placental MAOA gene expression. These effects could compromise optimal temperamental trajectory and contribute to risk for psychological problems. Placental epigenetic markers can contribute to a multidimensional model of early intervention for high-risk children.
Collapse
Affiliation(s)
- Patricia M Pehme
- Clinical Psychology Program, The Graduate Center (CUNY), New York, New York, USA.,Department of Psychology, Queens College (CUNY), New York, New York, USA
| | - Wei Zhang
- Clinical Psychology Program, The Graduate Center (CUNY), New York, New York, USA.,Department of Psychology, Queens College (CUNY), New York, New York, USA
| | - Jackie Finik
- Department of Psychology, Queens College (CUNY), New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,CUNY Graduate School of Public Health and Health Policy, New York, New York, USA
| | - Alexandra Pritchett
- Clinical Psychology Program, The Graduate Center (CUNY), New York, New York, USA.,Department of Psychology, Queens College (CUNY), New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica Buthmann
- Department of Psychology, Queens College (CUNY), New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Behavioral and Cognitive Neuroscience Program, The Graduate Center (CUNY), New York, New York, USA
| | - Kathryn Dana
- Clinical Psychology Program, The Graduate Center (CUNY), New York, New York, USA.,Department of Psychology, Queens College (CUNY), New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yoko Nomura
- Clinical Psychology Program, The Graduate Center (CUNY), New York, New York, USA.,Department of Psychology, Queens College (CUNY), New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,CUNY Graduate School of Public Health and Health Policy, New York, New York, USA.,Behavioral and Cognitive Neuroscience Program, The Graduate Center (CUNY), New York, New York, USA
| |
Collapse
|
131
|
Associations between maternal prenatal cortisol and fetal growth are specific to infant sex: findings from the Wirral Child Health and Development Study. J Dev Orig Health Dis 2018; 9:425-431. [PMID: 29631648 DOI: 10.1017/s2040174418000181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent findings highlight that there are prenatal risks for affective disorders that are mediated by glucocorticoid mechanisms, and may be specific to females. There is also evidence of sex differences in prenatal programming mechanisms and developmental psychopathology, whereby effects are in opposite directions in males and females. As birth weight is a risk for affective disorders, we sought to investigate whether maternal prenatal cortisol may have sex-specific effects on fetal growth. Participants were 241 mothers selected from the Wirral Child Health and Development Study (WCHADS) cohort (n=1233) using a psychosocial risk stratifier, so that responses could be weighted back to the general population. Mothers provided saliva samples, which were assayed for cortisol, at home over 2 days at 32 weeks gestation (on waking, 30-min post-waking and during the evening). Measures of infant birth weight (corrected for gestational age) were taken from hospital records. General population estimates of associations between variables were obtained using inverse probability weights. Maternal log of the area under the curve cortisol predicted infant birth weight in a sex-dependent manner (interaction term P=0.029). There was a positive and statistically significant association between prenatal cortisol in males, and a negative association in females that was not statistically significant. A sex interaction in the same direction was evident when using the waking (P=0.015), and 30-min post-waking (P=0.013) cortisol, but not the evening measure. There was no interaction between prenatal cortisol and sex to predict gestational age. Our findings add to an emerging literature that suggests that there may be sex-specific mechanisms that underpin fetal programming.
Collapse
|
132
|
Zhang W, Li Q, Deyssenroth M, Lambertini L, Finik J, Ham J, Huang Y, Tsuchiya KJ, Pehme P, Buthmann J, Yoshida S, Chen J, Nomura Y. Timing of prenatal exposure to trauma and altered placental expressions of hypothalamic-pituitary-adrenal axis genes and genes driving neurodevelopment. J Neuroendocrinol 2018; 30:e12581. [PMID: 29423924 PMCID: PMC5939590 DOI: 10.1111/jne.12581] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/30/2018] [Accepted: 02/03/2018] [Indexed: 12/17/2022]
Abstract
Prenatal maternal stress increases the risk for negative developmental outcomes in offspring; however, the underlying biological mechanisms remain largely unexplored. In the present study, alterations in placental gene expression associated with maternal stress were examined to clarify the potential underlying epi/genetic mechanisms. Expression levels of 40 selected genes involved in regulating foetal hypothalamic-pituitary-adrenal axis and neurodevelopment were profiled in placental tissues collected from a birth cohort established around the time of Superstorm Sandy. Objective prenatal traumatic stress was defined as whether mothers were exposed to Superstorm Sandy during pregnancy. Among the 275 mother-infant dyads, 181 dyads were delivered before Superstorm Sandy (ie, Control), 66 dyads were exposed to Superstorm Sandy during the first trimester (ie, Early Exposure) and 28 were exposed to Superstorm Sandy during the second or third trimester (ie, Mid-Late Exposure). Across all trimesters, expression of HSD11B2, MAOA, ZNF507 and DYRK1A was down-regulated among those exposed to Superstorm Sandy during pregnancy. Furthermore, trimester-specific differences were also observed: exposure during early gestation was associated with down-regulation of HSD11B1 and MAOB and up-regulation of CRHBP; exposure during mid-late gestation was associated with up-regulation of SRD5A3. The findings of the present study suggest that placental gene expression may be altered in response to traumatic stress exposure during pregnancy, and the susceptibility of these genes is dependent on the time of the exposure during pregnancy. Further studies should aim to clarify the biological mechanisms that underlie trimester-specific exposure by evaluating the differential impact on offspring neurodevelopment later in childhood.
Collapse
Affiliation(s)
- Wei Zhang
- Queens College, CUNY, Psychology, New York, NY, United States
| | - Qian Li
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Maya Deyssenroth
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Luca Lambertini
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Jackie Finik
- Queens College, CUNY, Psychology, New York, NY, United States
- The Graduate Center, CUNY, Psychology, New York, NY, United States
- Graduate School of Public Health and Health Policy, CUNY, New York, NY, United State
| | - Jacob Ham
- Icahn School of Medicine at Mount Sinai, Psychiatry, New York, NY, United States
| | - Yongling Huang
- The Graduate Center, CUNY, Psychology, New York, NY, United States
| | - Kenji J Tsuchiya
- Hamamatsu University School of Medicine, Research Center for Child Mental Development, Shizuoka, Japan
| | - Patricia Pehme
- Queens College, CUNY, Psychology, New York, NY, United States
- The Graduate Center, CUNY, Psychology, New York, NY, United States
| | - Jessica Buthmann
- Queens College, CUNY, Psychology, New York, NY, United States
- The Graduate Center, CUNY, Psychology, New York, NY, United States
| | - Sachiko Yoshida
- Department of Environmental & Life Sciences, Toyohashi University of Technology, Toyohashi, Japan
| | - Jia Chen
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Yoko Nomura
- Queens College, CUNY, Psychology, New York, NY, United States
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
- The Graduate Center, CUNY, Psychology, New York, NY, United States
- Icahn School of Medicine at Mount Sinai, Psychiatry, New York, NY, United States
| |
Collapse
|
133
|
Sheriff MJ, Bell A, Boonstra R, Dantzer B, Lavergne SG, McGhee KE, MacLeod KJ, Winandy L, Zimmer C, Love OP. Integrating Ecological and Evolutionary Context in the Study of Maternal Stress. Integr Comp Biol 2018; 57:437-449. [PMID: 28957523 DOI: 10.1093/icb/icx105] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Maternal stress can prenatally influence offspring phenotypes and there are an increasing number of ecological studies that are bringing to bear biomedical findings to natural systems. This is resulting in a shift from the perspective that maternal stress is unanimously costly, to one in which maternal stress may be beneficial to offspring. However, this adaptive perspective is in its infancy with much progress to still be made in understanding the role of maternal stress in natural systems. Our aim is to emphasize the importance of the ecological and evolutionary context within which adaptive hypotheses of maternal stress can be evaluated. We present five primary research areas where we think future research can make substantial progress: (1) understanding maternal and offspring control mechanisms that modulate exposure between maternal stress and subsequent offspring phenotype response; (2) understanding the dynamic nature of the interaction between mothers and their environment; (3) integrating offspring phenotypic responses and measuring both maternal and offspring fitness outcomes under real-life (either free-living or semi-natural) conditions; (4) empirically testing these fitness outcomes across relevant spatial and temporal environmental contexts (both pre- and post-natal environments); (5) examining the role of maternal stress effects in human-altered environments-i.e., do they limit or enhance fitness. To make progress, it is critical to understand the role of maternal stress in an ecological context and to do that, we must integrate across physiology, behavior, genetics, and evolution.
Collapse
Affiliation(s)
- Michael J Sheriff
- Department of Ecosystem Science and Management, Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Alison Bell
- School of Integrative Biology, Program in Neuroscience, and Program in Ecology, Evolution and Conservation Biology, Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, IL 61821, USA
| | - Rudy Boonstra
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Ben Dantzer
- Department of Psychology, and Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sophia G Lavergne
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Katie E McGhee
- Department of Biology, the University of the South, Sewanee, TN 37383, USA
| | - Kirsty J MacLeod
- Department of Ecosystem Science and Management, Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.,Department of Biology, Mueller Laboratory, Pennsylvania State University, University Park, PA 16802, USA
| | - Laurane Winandy
- CNRS, Université Toulouse 3 Paul Sabatier, ENFA, UMR5174 (Laboratoire Évolution and Diversité Biologique), 31077 Toulouse, France.,CNRS, UMR5321, Station d'Ecologie Théorique et Expérimentale, 09200 Moulis, France
| | - Cedric Zimmer
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853, USA
| | - Oliver P Love
- Department of Biological Sciences, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| |
Collapse
|
134
|
Tang L, He G, Liu X, Xu W. Progress in the understanding of the etiology and predictability of fetal growth restriction. Reproduction 2018; 153:R227-R240. [PMID: 28476912 DOI: 10.1530/rep-16-0287] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 02/21/2017] [Accepted: 03/14/2017] [Indexed: 12/12/2022]
Abstract
Fetal growth restriction (FGR) is defined as the failure of fetus to reach its growth potential for various reasons, leading to multiple perinatal complications and adult diseases of fetal origins. Shallow extravillous trophoblast (EVT) invasion-induced placental insufficiency and placental dysfunction are considered the main reasons for idiopathic FGR. In this review, first we discuss the major characteristics of anti-angiogenic state and the pro-inflammatory bias in FGR. We then elaborate major abnormalities in placental insufficiency at molecular levels, including the interaction between decidual leukocytes and EVT, alteration of miRNA expression and imprinted gene expression pattern in FGR. Finally, we review current animal models used in FGR, an experimental intervention based on animal models and the progress of predictive biomarker studies in FGR.Free Chinese abstract: A Chinese translation of this abstract is freely available at http://www.reproduction-online.org/content/153/6/R215/suppl/DC1.
Collapse
Affiliation(s)
- Li Tang
- Joint Laboratory of Reproductive MedicineSCU-CUHK, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education.,Department of Obstetric and Gynecologic DiseasesWest China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Guolin He
- Department of Obstetric and Gynecologic DiseasesWest China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinghui Liu
- Department of Obstetric and Gynecologic DiseasesWest China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wenming Xu
- Joint Laboratory of Reproductive MedicineSCU-CUHK, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education .,Department of Obstetric and Gynecologic DiseasesWest China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
135
|
Rock KD, Horman B, Phillips AL, McRitchie SL, Watson S, Deese-Spruill J, Jima D, Sumner S, Stapleton HM, Patisaul HB. EDC IMPACT: Molecular effects of developmental FM 550 exposure in Wistar rat placenta and fetal forebrain. Endocr Connect 2018; 7:305-324. [PMID: 29351906 PMCID: PMC5817967 DOI: 10.1530/ec-17-0373] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
Firemaster 550 (FM 550) is a flame retardant (FR) mixture that has become one of the most commonly used FRs in foam-based furniture and baby products. Human exposure to this commercial mixture, composed of brominated and organophosphate components, is widespread. We have repeatedly shown that developmental exposure can lead to sex-specific behavioral effects in rats. Accruing evidence of endocrine disruption and potential neurotoxicity has raised concerns regarding the neurodevelopmental effects of FM 550 exposure, but the specific mechanisms of action remains unclear. Additionally, we observed significant, and in some cases sex-specific, accumulation of FM 550 in placental tissue following gestational exposure. Because the placenta is an important source of hormones and neurotransmitters for the developing brain, it may be a critical target of toxicity to consider in the context of developmental neurotoxicity. Using a mixture of targeted and exploratory approaches, the goal of the present study was to identify possible mechanisms of action in the developing forebrain and placenta. Wistar rat dams were orally exposed to FM 550 (0, 300 or 1000 µg/day) for 10 days during gestation and placenta and fetal forebrain tissue collected for analysis. In placenta, evidence of endocrine, inflammatory and neurotransmitter signaling pathway disruption was identified. Notably, 5-HT turnover was reduced in placental tissue and fetal forebrains indicating that 5-HT signaling between the placenta and the embryonic brain may be disrupted. These findings demonstrate that environmental contaminants, like FM 550, have the potential to impact the developing brain by disrupting normal placental functions.
Collapse
Affiliation(s)
- Kylie D Rock
- Department of Biological SciencesNorth Carolina State University, Raleigh, North Carolina, USA
| | - Brian Horman
- Department of Biological SciencesNorth Carolina State University, Raleigh, North Carolina, USA
| | - Allison L Phillips
- Nicholas School of the EnvironmentDuke University, Durham, North Carolina, USA
| | - Susan L McRitchie
- NIH Eastern Regional Comprehensive Metabolomics Res. CoreUniv. of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Scott Watson
- NIH Eastern Regional Comprehensive Metabolomics Res. CoreUniv. of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jocelin Deese-Spruill
- NIH Eastern Regional Comprehensive Metabolomics Res. CoreUniv. of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dereje Jima
- Center for Human Health and the EnvironmentNorth Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research CenterNorth Carolina State University, Raleigh, North Carolina, USA
| | - Susan Sumner
- NIH Eastern Regional Comprehensive Metabolomics Res. CoreUniv. of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for Human Health and the EnvironmentNorth Carolina State University, Raleigh, North Carolina, USA
| | - Heather M Stapleton
- Nicholas School of the EnvironmentDuke University, Durham, North Carolina, USA
| | - Heather B Patisaul
- Department of Biological SciencesNorth Carolina State University, Raleigh, North Carolina, USA
- Center for Human Health and the EnvironmentNorth Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
136
|
Yehuda R, Meaney MJ. Relevance of Psychological Symptoms in Pregnancy to Intergenerational Effects of Preconception Trauma. Biol Psychiatry 2018; 83:94-96. [PMID: 29223220 DOI: 10.1016/j.biopsych.2017.10.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Michael J Meaney
- Sackler Program for Epigenetics and Psychobiology at McGill University, Montreal, Quebec, Canada; Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Singapore Institute for Clinical Sciences, Singapore
| |
Collapse
|
137
|
Abstract
Prenatal insults, such as maternal stress, are associated with an increased neurodevelopmental disease risk and impact males significantly more than females, including increased rates of autism, mental retardation, stuttering, dyslexia, and attention deficit/hyperactivity disorder (ADHD). Sex differences in the placenta, which begin with sex chromosomes, are likely to produce sex-specific transplacental signals to the developing brain. Our studies and others have identified X-linked genes that are expressed at higher levels in the female placenta. Through a genome-wide screen after maternal stress in mice, we identified the X-linked gene O-linked N-acetylglucosamine transferase (OGT) and demonstrated its causality in neurodevelopmental programming producing a male-specific stress phenotype. Elucidating the sex-specific molecular mechanisms involved in transplacental signals that impact brain development is key to understanding the sex bias in neurodevelopmental disorders and is expected to yield novel insight into disease risk and resilience.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine and Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
138
|
Haque M, Starr LM, Koski KG, Scott ME. Differential expression of genes in fetal brain as a consequence of maternal protein deficiency and nematode infection. Int J Parasitol 2017; 48:51-58. [PMID: 28903026 DOI: 10.1016/j.ijpara.2017.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/06/2017] [Accepted: 07/10/2017] [Indexed: 11/16/2022]
Abstract
Maternal dietary protein deficiency and gastrointestinal nematode infection during early pregnancy have negative impacts on both maternal placental gene expression and fetal growth in the mouse. Here we used next-generation RNA sequencing to test our hypothesis that maternal protein deficiency and/or nematode infection also alter the expression of genes in the developing fetal brain. Outbred pregnant CD1 mice were used in a 2×2 design with two levels of dietary protein (24% versus 6%) and two levels of infection (repeated sham versus Heligmosomoides bakeri beginning at gestation day 5). Pregnant dams were euthanized on gestation day 18 to harvest the whole fetal brain. Four fetal brains from each treatment group were analyzed using RNA Hi-Seq sequencing and the differential expression of genes was determined by the edgeR package using NetworkAnalyst. In response to maternal H. bakeri infection, 96 genes (88 up-regulated and eight down-regulated) were differentially expressed in the fetal brain. Differentially expressed genes were involved in metabolic processes, developmental processes and the immune system according to the PANTHER classification system. Among the important biological functions identified, several up-regulated genes have known neurological functions including neuro-development (Gdf15, Ing4), neural differentiation (miRNA let-7), synaptic plasticity (via suppression of NF-κβ), neuro-inflammation (S100A8, S100A9) and glucose metabolism (Tnnt1, Atf3). However, in response to maternal protein deficiency, brain-specific serine protease (Prss22) was the only up-regulated gene and only one gene (Dynlt1a) responded to the interaction of maternal nematode infection and protein deficiency. In conclusion, maternal exposure to GI nematode infection from day 5 to 18 of pregnancy may influence developmental programming of the fetal brain.
Collapse
Affiliation(s)
- Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Lisa M Starr
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada.
| |
Collapse
|
139
|
Baldwin KR, Phillips AL, Horman B, Arambula SE, Rebuli ME, Stapleton HM, Patisaul HB. Sex Specific Placental Accumulation and Behavioral Effects of Developmental Firemaster 550 Exposure in Wistar Rats. Sci Rep 2017; 7:7118. [PMID: 28769031 PMCID: PMC5540931 DOI: 10.1038/s41598-017-07216-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/23/2017] [Indexed: 12/20/2022] Open
Abstract
Firemaster® 550 (FM 550) is a commercial flame retardant mixture of brominated and organophosphate compounds applied to polyurethane foam used in furniture and baby products. Due to widespread human exposure, and structural similarities with known endocrine disruptors, concerns have been raised regarding possible toxicity. We previously reported evidence of sex specific behavioral effects in rats resulting from developmental exposure. The present study expands upon this prior finding by testing for a greater range of behavioral effects, and measuring the accumulation of FM 550 compounds in placental tissue. Wistar rat dams were orally exposed to FM 550 during gestation (0, 300 or 1000 µg/day; GD 9 – 18) for placental measurements or perinatally (0, 100, 300 or 1000 µg/day; GD 9 – PND 21) to assess activity and anxiety-like behaviors. Placental accumulation was dose dependent, and in some cases sex specific, with the brominated components reaching the highest levels. Behavioral changes were predominantly associated with a loss or reversal of sex differences in activity and anxiety-like behaviors. These findings demonstrate that environmental chemicals may sex-dependently accumulate in the placenta. That sex-biased exposure might translate to sex-specific adverse outcomes such as behavioral deficits is a possibility that merits further investigation.
Collapse
Affiliation(s)
- Kylie R Baldwin
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Allison L Phillips
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA
| | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Sheryl E Arambula
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Meghan E Rebuli
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | | | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA. .,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
140
|
Bandoli G, Palmsten K, Forbess Smith CJ, Chambers CD. A Review of Systemic Corticosteroid Use in Pregnancy and the Risk of Select Pregnancy and Birth Outcomes. Rheum Dis Clin North Am 2017; 43:489-502. [PMID: 28711148 PMCID: PMC5604866 DOI: 10.1016/j.rdc.2017.04.013] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The evidence to date regarding corticosteroid exposure in pregnancy and select pregnancy and birth outcomes is limited and inconsistent. The authors provide a narrative review of published literature summarizing the findings for oral clefts, preterm birth, birth weight, preeclampsia, and gestational diabetes mellitus. Whenever possible, the results are limited to oral or systemic administration with a further focus on use in autoimmune disease. Although previous studies of corticosteroid exposure in pregnancy reported an increased risk of oral clefts in the offspring, more recent studies have not replicated these findings.
Collapse
Affiliation(s)
- Gretchen Bandoli
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, Mail Code 0828, La Jolla, CA 92093-0412, USA.
| | - Kristin Palmsten
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, Mail Code 0828, La Jolla, CA 92093-0412, USA
| | - Chelsey J Forbess Smith
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, Mail Code 0656, La Jolla, CA 92093-0412, USA
| | - Christina D Chambers
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, Mail Code 0828, La Jolla, CA 92093-0412, USA
| |
Collapse
|
141
|
Miller GE, Borders AE, Crockett AH, Ross KM, Qadir S, Keenan-Devlin L, Leigh AK, Ham P, Ma J, Arevalo JM, Ernst LM, Cole SW. Maternal socioeconomic disadvantage is associated with transcriptional indications of greater immune activation and slower tissue maturation in placental biopsies and newborn cord blood. Brain Behav Immun 2017; 64:276-284. [PMID: 28434870 PMCID: PMC5493326 DOI: 10.1016/j.bbi.2017.04.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
Children from economically disadvantaged families experience worse cognitive, psychiatric, and medical outcomes compared to more affluent youth. Preclinical models suggest some of the adverse influence of disadvantage could be transmitted during gestation via maternal immune activation, but this hypothesis has not been tested in humans. It also remains unclear whether prenatal interventions can mitigate such effects. To fill these gaps, we conducted two studies. Study 1 characterized the socioeconomic conditions of 79 women during pregnancy. At delivery, placenta biopsies and umbilical blood were collected for transcriptional profiling. Maternal disadvantage was associated with a transcriptional profile indicative of higher immune activation and slower fetal maturation, particularly in pathways related to brain, heart, and immune development. Cord blood cells of disadvantaged newborns also showed indications of immaturity, as reflected in down-regulation of pathways that coordinate myeloid cell development. These associations were independent of fetal sex, and characteristics of mothers (age, race, adiposity, diabetes, pre-eclampsia) and babies (delivery method, gestational age). Study 2 performed the same transcriptional analyses in specimens from 20 women participating in CenteringPregnancy, a group-based psychosocial intervention, and 20 women in traditional prenatal care. In both placenta biopsies and cord blood, women in CenteringPregnancy showed up-regulation of transcripts found in Study 1 to be most down-regulated in conjunction with disadvantage. Collectively, these results suggest socioeconomic disparities in placental biology are evident at birth, and provide clues about the mechanistic origins of health disparities. They also suggest the possibility that psychosocial interventions could have mitigating influences.
Collapse
Affiliation(s)
- Gregory E. Miller
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Ann E. Borders
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Amy H. Crockett
- Department of Obstetrics & Gynecology, Greenville Hospital System University Medical Center, Greenville SC
| | - Kharah M. Ross
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Sameen Qadir
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Lauren Keenan-Devlin
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Adam K. Leigh
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Paula Ham
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Jeffrey Ma
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| | - Jesusa M.G. Arevalo
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| | - Linda M. Ernst
- Department of Pathology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Steve W. Cole
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| |
Collapse
|
142
|
Bronson SL, Chan JC, Bale TL. Sex-Specific Neurodevelopmental Programming by Placental Insulin Receptors on Stress Reactivity and Sensorimotor Gating. Biol Psychiatry 2017; 82:127-138. [PMID: 28168960 PMCID: PMC5483189 DOI: 10.1016/j.biopsych.2016.12.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/22/2016] [Accepted: 12/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diabetes, obesity, and overweight are prevalent pregnancy complications that predispose offspring to neurodevelopmental disorders, including autism, attention-deficit/hyperactivity disorder, and schizophrenia. Although male individuals are three to four times more likely than female individuals to develop these disorders, the mechanisms driving the sex specificity of disease vulnerability remain unclear. Because defective placental insulin receptor (InsR) signaling is a hallmark of pregnancy metabolic dysfunction, we hypothesized that it may be an important contributor and novel mechanistic link to sex-specific neurodevelopmental changes underlying disease risk. METHODS We used Cre/loxP transgenic mice to conditionally target InsRs in fetally derived placental trophoblasts. Adult offspring were evaluated for effects of placental trophoblast-specific InsR deficiency on stress sensitivity, cognitive function, sensorimotor gating, and prefrontal cortical transcriptional reprogramming. To evaluate molecular mechanisms driving sex-specific outcomes, we assessed genome-wide expression profiles in the placenta and fetal brain. RESULTS Male, but not female, mice with placental trophoblast-specific InsR deficiency showed a significantly increased hypothalamic-pituitary-adrenal axis stress response and impaired sensorimotor gating, phenotypic effects that were associated with dysregulated nucleotide metabolic processes in the male prefrontal cortex. Within the placenta, InsR deficiency elicited changes in gene expression, predominantly in male mice, reflecting potential shifts in vasculature, amino acid transport, serotonin homeostasis, and mitochondrial function. These placental disruptions were associated with altered gene expression profiles in the male fetal brain and suggested delayed cortical development. CONCLUSIONS Together, these data demonstrate the novel role of placental InsRs in sex-specific neurodevelopment and reveal a potential mechanism for neurodevelopmental disorder risk in pregnancies complicated by maternal metabolic disorders, including diabetes and obesity.
Collapse
Affiliation(s)
- Stefanie L Bronson
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer C Chan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
143
|
Varcin KJ, Alvares GA, Uljarević M, Whitehouse AJO. Prenatal maternal stress events and phenotypic outcomes in Autism Spectrum Disorder. Autism Res 2017; 10:1866-1877. [DOI: 10.1002/aur.1830] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/20/2017] [Accepted: 06/05/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Kandice J. Varcin
- Telethon Kids Institute, University of Western Australia; Perth Western Australia Australia
| | - Gail A. Alvares
- Telethon Kids Institute, University of Western Australia; Perth Western Australia Australia
- Cooperative Research Centre for Living with Autism (Autism CRC); Long Pocket Brisbane, Queensland Australia
| | - Mirko Uljarević
- Cooperative Research Centre for Living with Autism (Autism CRC); Long Pocket Brisbane, Queensland Australia
- Olga Tennison Autism Research Centre; School of Psychology and Public Health, La Trobe University; Victoria Australia
| | | |
Collapse
|
144
|
Socioeconomic disadvantage, gestational immune activity, and neurodevelopment in early childhood. Proc Natl Acad Sci U S A 2017; 114:6728-6733. [PMID: 28607066 DOI: 10.1073/pnas.1617698114] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Children raised in economically disadvantaged households face increased risks of poor health in adulthood, suggesting that inequalities in health have early origins. From the child's perspective, exposure to economic hardship may begin as early as conception, potentially via maternal neuroendocrine-immune responses to prenatal stressors, which adversely impact neurodevelopment. Here we investigate whether socioeconomic disadvantage is associated with gestational immune activity and whether such activity is associated with abnormalities among offspring during infancy. We analyzed concentrations of five immune markers (IL-1β, IL-6, IL-8, IL-10, and TNF-α) in maternal serum from 1,494 participants in the New England Family Study in relation to the level of maternal socioeconomic disadvantage and their involvement in offspring neurologic abnormalities at 4 mo and 1 y of age. Median concentrations of IL-8 were lower in the most disadvantaged pregnancies [-1.53 log(pg/mL); 95% CI: -1.81, -1.25]. Offspring of these pregnancies had significantly higher risk of neurologic abnormalities at 4 mo [odds ratio (OR) = 4.61; CI = 2.84, 7.48] and 1 y (OR = 2.05; CI = 1.08, 3.90). This higher risk was accounted for in part by fetal exposure to lower maternal IL-8, which also predicted higher risks of neurologic abnormalities at 4 mo (OR = 7.67; CI = 4.05, 14.49) and 1 y (OR = 2.92; CI = 1.46, 5.87). Findings support the role of maternal immune activity in fetal neurodevelopment, exacerbated in part by socioeconomic disadvantage. This finding reveals a potential pathophysiologic pathway involved in the intergenerational transmission of socioeconomic inequalities in health.
Collapse
|
145
|
Maternal and Early-Life Circadian Disruption Have Long-Lasting Negative Consequences on Offspring Development and Adult Behavior in Mice. Sci Rep 2017; 7:3326. [PMID: 28607386 PMCID: PMC5468226 DOI: 10.1038/s41598-017-03406-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/28/2017] [Indexed: 01/31/2023] Open
Abstract
Modern life involves chronic circadian disruption through artificial light and these disruptions are associated with numerous mental and physical health maladies. Because the developing nervous system is particularly vulnerable to perturbation, we hypothesized that early-life circadian disruption would negatively impact offspring development and adult function. Pregnant mice were subjected to chronic circadian disruption from the time of uterine implantation through weaning. To dissociate in utero from postnatal effects, a subset of litters was cross-fostered at birth from disrupted dams to control dams and vice versa. Postnatal circadian disruption was associated with reduced adult body mass, social avoidance, and hyperactivity. In utero disruption resulted in more pronounced social avoidance and hyperactivity, phenotypes not abrogated by cross-fostering to control mothers. To examine whether circadian disruption affects development by acting as an early life stressor, we examined birthweight, litter size, maternal cannibalism, and epigenetic modifications. None of these variables differed between control and disrupted dams, or resembled patterns seen following early-life stress. Our findings indicate that developmental chronic circadian disruption permanently affects somatic and behavioral development in a stage-of-life-dependent manner, independent of early life stress mechanisms, underscoring the importance of temporal structure during development, both in utero and early postnatal life.
Collapse
|
146
|
Abstract
Epidemiological evidence links an individual's susceptibility to chronic disease in adult life to events during their intrauterine phase of development. Biologically this should not be unexpected, for organ systems are at their most plastic when progenitor cells are proliferating and differentiating. Influences operating at this time can permanently affect their structure and functional capacity, and the activity of enzyme systems and endocrine axes. It is now appreciated that such effects lay the foundations for a diverse array of diseases that become manifest many years later, often in response to secondary environmental stressors. Fetal development is underpinned by the placenta, the organ that forms the interface between the fetus and its mother. All nutrients and oxygen reaching the fetus must pass through this organ. The placenta also has major endocrine functions, orchestrating maternal adaptations to pregnancy and mobilizing resources for fetal use. In addition, it acts as a selective barrier, creating a protective milieu by minimizing exposure of the fetus to maternal hormones, such as glucocorticoids, xenobiotics, pathogens, and parasites. The placenta shows a remarkable capacity to adapt to adverse environmental cues and lessen their impact on the fetus. However, if placental function is impaired, or its capacity to adapt is exceeded, then fetal development may be compromised. Here, we explore the complex relationships between the placental phenotype and developmental programming of chronic disease in the offspring. Ensuring optimal placentation offers a new approach to the prevention of disorders such as cardiovascular disease, diabetes, and obesity, which are reaching epidemic proportions.
Collapse
Affiliation(s)
- Graham J Burton
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Abigail L Fowden
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Kent L Thornburg
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
147
|
Chin EH, Schmidt KL, Martel KM, Wong CK, Hamden JE, Gibson WT, Soma KK, Christians JK. A maternal high-fat, high-sucrose diet has sex-specific effects on fetal glucocorticoids with little consequence for offspring metabolism and voluntary locomotor activity in mice. PLoS One 2017; 12:e0174030. [PMID: 28301585 PMCID: PMC5354465 DOI: 10.1371/journal.pone.0174030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 03/02/2017] [Indexed: 01/21/2023] Open
Abstract
Maternal overnutrition and obesity during pregnancy can have long-term effects on offspring physiology and behaviour. These developmental programming effects may be mediated by fetal exposure to glucocorticoids, which is regulated in part by placental 11β-hydroxysteroid dehydrogenase (11β-HSD) type 1 and 2. We tested whether a maternal high-fat, high-sucrose diet would alter expression of placental 11β-HSD1 and 2, thereby increasing fetal exposure to maternal glucocorticoids, with downstream effects on offspring physiology and behaviour. C57BL/6J mice were fed a high-fat, high-sucrose (HFHS) diet or a nutrient-matched low-fat, no-sucrose control diet prior to and during pregnancy and lactation. At day 17 of gestation, HFHS dams had ~20% lower circulating corticosterone levels than controls. Furthermore, there was a significant interaction between maternal diet and fetal sex for circulating corticosterone levels in the fetuses, whereby HFHS males tended to have higher corticosterone than control males, with no effect in female fetuses. However, placental 11β-HSD1 or 11β-HSD2 expression did not differ between diets or show an interaction between diet and sex. To assess potential long-term consequences of this sex-specific effect on fetal corticosterone, we studied locomotor activity and metabolic traits in adult offspring. Despite a sex-specific effect of maternal diet on fetal glucocorticoids, there was little evidence of sex-specific effects on offspring physiology or behaviour, although HFHS offspring of both sexes had higher circulating corticosterone at 9 weeks of age. Our results suggest the existence of as yet unknown mechanisms that mitigate the effects of altered glucocorticoid exposure early in development, making offspring resilient to the potentially negative effects of a HFHS maternal diet.
Collapse
Affiliation(s)
- Eunice H. Chin
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Kim L. Schmidt
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Kaitlyn M. Martel
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Chi Kin Wong
- Department of Medical Genetics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Jordan E. Hamden
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - William T. Gibson
- Department of Medical Genetics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Kiran K. Soma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Julian K. Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- * E-mail:
| |
Collapse
|
148
|
Martin E, Smeester L, Bommarito PA, Grace MR, Boggess K, Kuban K, Karagas MR, Marsit CJ, O’Shea TM, Fry RC. Sexual epigenetic dimorphism in the human placenta: implications for susceptibility during the prenatal period. Epigenomics 2017; 9:267-278. [PMID: 28234023 PMCID: PMC5331919 DOI: 10.2217/epi-2016-0132] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
AIM Sex-based differences in response to adverse prenatal environments and infant outcomes have been observed, yet the underlying mechanisms for this are unclear. The placental epigenome may be a driver of these differences. METHODS Placental DNA methylation was assessed at more than 480,000 CpG sites from male and female infants enrolled in the extremely low gestational age newborns cohort (ELGAN) and validated in a separate US-based cohort. The impact of gestational age on placental DNA methylation was further examined using the New Hampshire Birth Cohort Study for a total of n = 467 placentas. RESULTS A total of n = 2745 CpG sites, representing n = 587 genes, were identified as differentially methylated (p < 1 × 10-7). The majority (n = 582 or 99%) of these were conserved among the New Hampshire Birth Cohort. The identified genes encode proteins related to immune function, growth/transcription factor signaling and transport across cell membranes. CONCLUSION These data highlight sex-dependent epigenetic patterning in the placenta and provide insight into differences in infant outcomes and responses to the perinatal environment.
Collapse
Affiliation(s)
- Elizabeth Martin
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Lisa Smeester
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Paige A Bommarito
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Matthew R Grace
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Kim Boggess
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Karl Kuban
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA 30322, USA
| | - T Michael O’Shea
- Department of Pediatrics, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
149
|
Delunardo F, Soldati D, Bellisario V, Berry A, Camerini S, Crescenzi M, Alessandri C, Conti F, Ceccarelli F, Francia A, Valesini G, Cirulli F, Siracusano A, Siracusano A, Niolu C, Alex Rubino I, Ortona E, Margutti P. Anti-GAPDH Autoantibodies as a Pathogenic Determinant and Potential Biomarker of Neuropsychiatric Diseases. Arthritis Rheumatol 2016; 68:2708-2716. [PMID: 27213890 DOI: 10.1002/art.39750] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/05/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the potential role of circulating autoantibodies specific to neuronal cell surface antigens in the pathophysiology of neuropsychiatric disorders. METHODS Two different kinds of immunoscreening approaches were used to identify autoantigens associated with neuropsychiatric disorders in the serum of patients with schizophrenia. The presence of autoantibodies specific to the identified autoantigens was then tested in patients with various psychiatric disorders and in patients with systemic lupus erythematosus (SLE) and concomitant neuropsychiatric manifestations. Furthermore, the potential pathogenic role of these autoantibodies was assessed both in vitro and in vivo. RESULTS GAPDH was identified as a novel autoantigen associated with neuropsychiatric disorders. Serum anti-GAPDH IgG was detected in the serum of 51% of patients with schizophrenia and 50% of patients with major depression. Moreover, SLE patients with comorbid psychiatric manifestations presented significantly higher serum levels of anti-GAPDH antibodies than did SLE patients without psychiatric manifestations (P = 0.004 by chi-square test). Of note, a significant positive correlation (R = 0.48, P = 0.0049, by Spearman's rank correlation test) was found between the levels of serum anti-GAPDH antibodies and cognitive dysfunction in patients with SLE. In vitro analysis of the effects of purified human anti-GAPDH autoantibodies on SH-SY5Y cells showed an immediate neurite retraction. Finally, in vivo administration of anti-GAPDH autoantibodies in the right cerebral ventricle of C57BL/6J mice resulted in specific behavioral changes associated with a detrimental cognitive and emotional profile. CONCLUSION Overall, these data suggest that anti-GAPDH autoantibodies play a role in the pathogenesis of neuropsychiatric disorders, thus representing a potentially promising tool for the screening of individual vulnerability to these disabling conditions.
Collapse
|
150
|
Spinello C, Laviola G, Macrì S. Pediatric Autoimmune Disorders Associated with Streptococcal Infections and Tourette's Syndrome in Preclinical Studies. Front Neurosci 2016; 10:310. [PMID: 27445678 PMCID: PMC4928151 DOI: 10.3389/fnins.2016.00310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/20/2016] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence suggests that Tourette's Syndrome (TS) - a multifactorial pediatric disorder characterized by the recurrent exhibition of motor tics and/or vocal utterances - can partly depend on immune dysregulation provoked by early repeated streptococcal infections. The natural and adaptive antibody-mediated reaction to streptococcus has been proposed to potentially turn into a pathological autoimmune response in vulnerable individuals. Specifically, in conditions of increased permeability of the blood brain barrier (BBB), streptococcus-induced antibodies have been proposed to: (i) reach neuronal targets located in brain areas responsible for motion control; and (ii) contribute to the exhibition of symptoms. This theoretical framework is supported by indirect evidence indicating that a subset of TS patients exhibit elevated streptococcal antibody titers upon tic relapses. A systematic evaluation of this hypothesis entails preclinical studies providing a proof of concept of the aforementioned pathological sequelae. These studies shall rest upon individuals characterized by a vulnerable immune system, repeatedly exposed to streptococcus, and carefully screened for phenotypes isomorphic to the pathological signs of TS observed in patients. Preclinical animal models may thus constitute an informative, useful tool upon which conducting targeted, hypothesis-driven experiments. In the present review we discuss the available evidence in preclinical models in support of the link between TS and pediatric autoimmune neuropsychiatric disorders associated with streptococcus infections (PANDAS), and the existing gaps that future research shall bridge. Specifically, we report recent preclinical evidence indicating that the immune responses to repeated streptococcal immunizations relate to the occurrence of behavioral and neurological phenotypes reminiscent of TS. By the same token, we discuss the limitations of these studies: limited evidence of behavioral phenotypes isomorphic to tics and scarce knowledge about the immunological phenomena favoring the transition from natural adaptive immunity to pathological outcomes.
Collapse
Affiliation(s)
- Chiara Spinello
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Giovanni Laviola
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Simone Macrì
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| |
Collapse
|