1
|
Jean-Louis A, Gleason JL, Chen Z, Wagner KA, Grobman WA, Newman RB, Lee W, Gore-Langton R, Sherman S, Owen J, Wing DA, Skupski DW, Grewal J, Grantz KL. Fetal Body Composition and Organ Volume Trajectories in Association With Maternal Perceived Stress or Depressive Symptoms in the Fetal 3D Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025; 44:1245-1259. [PMID: 40129188 DOI: 10.1002/jum.16680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVES Evaluate associations between maternal perceived stress and depressive symptoms, and fetal body composition and organ volumes. METHODS In the NICHD Fetal 3D Study (2015-2019; n = 2457), stress and depressive symptoms were assessed at enrollment (10-13 weeks). High stress was defined as a Perceived Stress Scale (PSS) score ≥15 and high depressive symptoms, defined as an Edinburgh Postnatal Depression Scale (EPDS) score ≥10. Fetal body composition and organ volumes (cerebellar, lung, liver, and kidney) were measured up to five times between 15 and 40 weeks using three-dimensional (3D) ultrasound technology. Trajectories of body composition and organ volumes across pregnancy by high versus low PSS and EPDS were created using linear mixed models. Overall and weekly differences were compared after adjusting for covariates. RESULTS Six hundred (24.4%) women scored ≥15 on the PSS, and 334 (13.6%) scored ≥10 on the EPDS. Fetuses whose mothers had PSS ≥15 had 0.4-0.9 cm3 smaller fractional fat arm volumes between 33 and 37 weeks, 0.2-0.5 cm3 larger fractional fat thigh volumes between 26 and 29 weeks, 4.0-8.0 cm3 larger average lung volumes between 33 and 37 weeks, and 1.0 cm3 smaller liver volumes between 19 and 21 weeks; and for EPDS ≥10, fetuses had 2.0-3.0 cm3 larger cerebellar volumes between 35 and 38 weeks. Findings were statistically significant (P < .05) and similarly robust in sensitivity analyses. CONCLUSIONS Greater maternal perceived stress and depressive symptoms early in pregnancy were associated with up to an 8.0 cm3 difference in fetal limb adiposity and organ volume growth. While health implications are unclear, our findings support current clinical practice guidelines for prenatal mental health screening.
Collapse
Affiliation(s)
- Alexandra Jean-Louis
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica L Gleason
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhen Chen
- Biostatistics and Bioinformatics Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathryn A Wagner
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - William A Grobman
- Department of Obstetrics and Gynecology, Brown University Medical School, Providence, Rhode Island, USA
| | - Roger B Newman
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wesley Lee
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - John Owen
- Department of Obstetrics and Gynecology, Maternal-Fetal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Deborah A Wing
- Division of Obstetrics & Gynecology and Maternal-Fetal Medicine, University of California Irvine, Irvine, California, USA
| | - Daniel W Skupski
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, New York, USA
| | - Jagteshwar Grewal
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Frueh L, Sharma R, Sheffield PE, Clougherty JE. Community violence and asthma: A review. Ann Allergy Asthma Immunol 2024; 133:641-648.e12. [PMID: 39038705 DOI: 10.1016/j.anai.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Over the past 2 decades, epidemiologic studies have identified significant associations between exposure to violence, as a psychosocial stressor, and the incidence or exacerbation of asthma. Across diverse populations, study designs, and measures of community violence, researchers have consistently identified adverse associations. In this review, the published epidemiologic evidence is summarized with special attention to research published in the last 5 years and seminal papers. Hypothesized mechanisms for the direct effects of violence exposure and for how such exposure affects susceptibility to physical agents (eg, air pollution and extreme temperature) are discussed. These include stress-related pathways, behavioral mechanisms, and epigenetic mechanisms. Finally, clinical implications and recommendations are discussed.
Collapse
Affiliation(s)
- Lisa Frueh
- Department of Environmental and Occupational Health, Drexel University Dornsife School of Public Health, Philadelphia, Pennsylvania.
| | - Rachit Sharma
- Department of Environmental and Occupational Health, Drexel University Dornsife School of Public Health, Philadelphia, Pennsylvania
| | - Perry E Sheffield
- Departments of Environmental Medicine and Climate Science and Public Health and Pediatrics, Mount Sinai Icahn School of Medicine, New York, New York
| | - Jane E Clougherty
- Department of Environmental and Occupational Health, Drexel University Dornsife School of Public Health, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Lebold KM, Cook M, Pincus AB, Nevonen KA, Davis BA, Carbone L, Calco GN, Pierce AB, Proskocil BJ, Fryer AD, Jacoby DB, Drake MG. Grandmaternal allergen sensitization reprograms epigenetic and airway responses to allergen in second-generation offspring. Am J Physiol Lung Cell Mol Physiol 2023; 325:L776-L787. [PMID: 37814791 PMCID: PMC11068409 DOI: 10.1152/ajplung.00103.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023] Open
Abstract
Asthma susceptibility is influenced by environmental, genetic, and epigenetic factors. DNA methylation is one form of epigenetic modification that regulates gene expression and is both inherited and modified by environmental exposures throughout life. Prenatal development is a particularly vulnerable time period during which exposure to maternal asthma increases asthma risk in offspring. How maternal asthma affects DNA methylation in offspring and what the consequences of differential methylation are in subsequent generations are not fully known. In this study, we tested the effects of grandmaternal house dust mite (HDM) allergen sensitization during pregnancy on airway physiology and inflammation in HDM-sensitized and challenged second-generation mice. We also tested the effects of grandmaternal HDM sensitization on tissue-specific DNA methylation in allergen-naïve and -sensitized second-generation mice. Descendants of both allergen- and vehicle-exposed grandmaternal founders exhibited airway hyperreactivity after HDM sensitization. However, grandmaternal allergen sensitization significantly potentiated airway hyperreactivity and altered the epigenomic trajectory in second-generation offspring after HDM sensitization compared with HDM-sensitized offspring from vehicle-exposed founders. As a result, biological processes and signaling pathways associated with epigenetic modifications were distinct between lineages. A targeted analysis of pathway-associated gene expression found that Smad3 was significantly dysregulated as a result of grandmaternal allergen sensitization. These data show that grandmaternal allergen exposure during pregnancy establishes a unique epigenetic trajectory that reprograms allergen responses in second-generation offspring and may contribute to asthma risk.NEW & NOTEWORTHY Asthma susceptibility is influenced by environmental, genetic, and epigenetic factors. This study shows that maternal allergen exposure during pregnancy promotes unique epigenetic trajectories in second-generation offspring at baseline and in response to allergen sensitization, which is associated with the potentiation of airway hyperreactivity. These effects are one mechanism by which maternal asthma may influence the inheritance of asthma risk.
Collapse
Affiliation(s)
- Katie M Lebold
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, California, United States
| | - Madeline Cook
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Alexandra B Pincus
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Kimberly A Nevonen
- Knight Cardiovascular Institute Epigenetics Consortium, Oregon Health and Science University, Portland, Oregon, United States
| | - Brett A Davis
- Knight Cardiovascular Institute Epigenetics Consortium, Oregon Health and Science University, Portland, Oregon, United States
| | - Lucia Carbone
- Knight Cardiovascular Institute Epigenetics Consortium, Oregon Health and Science University, Portland, Oregon, United States
- Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, Oregon, United States
| | - Gina N Calco
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Aubrey B Pierce
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Becky J Proskocil
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Allison D Fryer
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - David B Jacoby
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Matthew G Drake
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
4
|
Freeman RE, Qi YS, Geller RJ, Cohen AR, Iyer SS, Waynik IY, Sullivan AF, Camargo CA. Parental Mental Health and Childhood Respiratory Outcomes in a Severe Bronchiolitis Cohort. Clin Pediatr (Phila) 2023; 62:1067-1079. [PMID: 36715245 DOI: 10.1177/00099228221150608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Parental mental health conditions appear to contribute to the development of childhood respiratory illness. We investigated the relationship between parental mental health and childhood respiratory illness using data from a 17-center prospective cohort study of infants hospitalized with bronchiolitis between 2011 and 2014 (n = 921). Among 779 (84.6%) participants with self-reported parental mental health history data, 184 (23.6%) had parental history of depression and 155 (19.9%) had anxiety. Multivariable analyses found that both parental history of depression (hazard ratio [HR] 1.41, 95% confidence interval [CI] 1.01-1.99) and anxiety (HR 1.28, 95% CI 1.08-1.52) were associated with an increased risk of recurrent wheezing by age 3 years. However, only parental history of anxiety was associated with asthma by age 5 years (odds ratio 1.79, 95% CI 1.25-2.55). Further research on exposure severity, other early life stressors, and effective methods of parental psychosocial support is needed to develop targeted risk factor prevention strategies to reduce the burden of childhood respiratory illness.
Collapse
Affiliation(s)
- Rain E Freeman
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Population Health Research, School of Public and Community Health Sciences, University of Montana, Missoula, MT, USA
| | - Ying Shelly Qi
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruth J Geller
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ari R Cohen
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sujit S Iyer
- Department of Pediatric Emergency Medicine, Dell Children's Medical Center of Central Texas, Austin, TX, USA
| | - Ilana Y Waynik
- Connecticut Children's Medical Center, Hartford, CT, USA
| | - Ashley F Sullivan
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Yadav A, Pacheco SE. Prebirth effects of climate change on children's respiratory health. Curr Opin Pediatr 2023; 35:344-349. [PMID: 36974440 DOI: 10.1097/mop.0000000000001241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
PURPOSE OF REVIEW To date, there is no evidence that humanity will implement appropriate mitigation measures to avoid the catastrophic impact of climate change on the planet and human health. Vulnerable populations such as pregnant women and children will be the most affected. This review highlights epidemiologic data on climate change-related prenatal environmental exposures affecting the fetus and children's respiratory health. RECENT FINDINGS Research on outcomes of prenatal exposure to climate change-related environmental changes and pediatric pulmonary health is limited. In addition to adverse pregnancy outcomes known to affect lung development, changes in lung function, increased prevalence of wheezing, atopy, and respiratory infections have been associated with prenatal exposure to increased temperatures, air pollution, and maternal stress. The mechanisms behind these changes are ill-defined, although oxidative stress, impaired placental functioning, and epigenetic modifications have been observed. However, the long-term impact of these changes remains unknown. SUMMARY The detrimental impact of the climate crisis on pediatric respiratory health begins before birth, highlighting the inherent vulnerability of pregnant women and children. Research and advocacy, along with mitigation and adaptation measures, must be implemented to protect pregnant women and children, the most affected but the least responsible for the climate crisis.
Collapse
Affiliation(s)
- Aravind Yadav
- Division of Pulmonary Medicine, Department of Pediatrics, The University of Texas Health Science Center, McGovern Medical School, Houston, Texas, USA
| | | |
Collapse
|
6
|
Clausing ES, Tomlinson CJ, Non AL. Epigenetics and social inequalities in asthma and allergy. J Allergy Clin Immunol 2023:S0091-6749(23)00213-0. [PMID: 36804452 DOI: 10.1016/j.jaci.2023.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 02/21/2023]
Affiliation(s)
- Elizabeth S Clausing
- School of Global Integrative Studies, Department of Anthropology, University of Nebraska-Lincoln, Lincoln, Neb; Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln, Lincoln, Neb; Rural Drug Addiction Research Center, University of Nebraska-Lincoln, Lincoln, Neb
| | - Cassidy J Tomlinson
- Department of Anthropology, University of California San Diego, San Diego, Calif
| | - Amy L Non
- Department of Anthropology, University of California San Diego, San Diego, Calif.
| |
Collapse
|
7
|
Shi YY, Wei Q, Ma X, Zhang Y, Wang L, Shi HJ. Maternal affective and stress-related factors during pregnancy affect the occurrence of childhood allergic diseases: A Shanghai MCPC study. J Psychosom Res 2023; 165:111142. [PMID: 36630818 DOI: 10.1016/j.jpsychores.2022.111142] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To investigate the association between exposures to maternal affective and stress-related factors during pregnancy and allergies in children from birth to 2 years of age. METHODS We enrolled a total of 4178 children from the Shanghai Maternal-Child Pairs Cohort and measured maternal stress, anxiety, and depression during pregnancy by applying the Life Events Scale for Pregnant Women, Self-Rating Anxiety Scale, and the Center for Epidemiologic Studies-Depression Scale, respectively. Children's allergies were assessed by community physicians at 2, 6, 12, and 24 months, respectively; these included eczema, atopic dermatitis, food allergy, wheezing, asthma, and atopic rhinitis. We applied a latent class analysis (LCA) to these factors and analyzed the impacts of maternal affective and stress-related factors on childhood allergies by exploiting multivariate logistic regression. RESULTS Three distinct classes of children were revealed by LCA: healthy (79.8%), transient allergy (15.2%), and persistent allergy (4.9%). High maternal stress in both early and late pregnancy was associated with an increased risk of infant eczema at 2 months (aOR = 1.30, 95% CI = 1.01-1.67; aOR = 1.64, 95% CI = 1.14-2.36). Moreover, high maternal stress in late pregnancy was also associated with food allergy at 6 months, rhinitis at 2 years of age, and persistent allergy (aOR = 3.22, 95% CI = 1.27-8.12; aOR = 1.78, 95% CI = 1.01-3.15; and aOR = 1.93, 95% CI = 1.10-3.40). CONCLUSIONS The associations of maternal affective and stress-related factors during pregnancy with childhood allergies may vary by type and disease onset. We postulate that maternal stress in late pregnancy may exert a sustained negative effect on early childhood allergic diseases.
Collapse
Affiliation(s)
- Yu-Yang Shi
- Department of Maternal, Child and Adolescent Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Qian Wei
- Department of Maternal, Child and Adolescent Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xuemei Ma
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Yunhui Zhang
- Department of Environment Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Ling Wang
- Shanghai Medical College of Fudan University, Shanghai 200032, China.
| | - Hui-Jing Shi
- Department of Maternal, Child and Adolescent Health, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW To provide an update on the current understanding of the role of wingless/integrase-1 (Wnt) signaling in pediatric allergic asthma and other pediatric lung diseases. RECENT FINDINGS The Wnt signaling pathway is critical for normal lung development. Genetic and epigenetic human studies indicate a link between Wnt signaling and the development and severity of asthma in children. Mechanistic studies using animal models of allergic asthma demonstrate a key role for Wnt signaling in allergic airway inflammation and remodeling. More recently, data on bronchopulmonary dysplasia (BPD) pathogenesis points to the Wnt signaling pathway as an important regulator. SUMMARY Current data indicates that the Wnt signaling pathway is an important mediator in allergic asthma and BPD pathogenesis. Further studies are needed to characterize the roles of individual Wnt signals in childhood disease, and to identify potential novel therapeutic targets to slow or prevent disease processes.
Collapse
Affiliation(s)
- Nooralam Rai
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Jeanine D’Armiento
- Department of Anesthesiology, Medicine, and Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
9
|
Haas-Neill S, Dvorkin-Gheva A, Forsythe P. Severe, but not moderate asthmatics share blood transcriptomic changes with post-traumatic stress disorder and depression. PLoS One 2022; 17:e0275864. [PMID: 36206293 PMCID: PMC9543640 DOI: 10.1371/journal.pone.0275864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
Abstract
Asthma, an inflammatory disorder of the airways, is one of the most common chronic illnesses worldwide and is associated with significant morbidity. There is growing recognition of an association between asthma and mood disorders including post-traumatic stress disorder (PTSD) and major depressive disorder (MDD). Although there are several hypotheses regarding the relationship between asthma and mental health, there is little understanding of underlying mechanisms and causality. In the current study we utilized publicly available datasets of human blood mRNA collected from patients with severe and moderate asthma, MDD, and PTSD. We performed differential expression (DE) analysis and Gene Set Enrichment Analysis (GSEA) on diseased subjects against the healthy subjects from their respective datasets, compared the results between diseases, and validated DE genes and gene sets with 4 more independent datasets. Our analysis revealed that commonalities in blood transcriptomic changes were only found between the severe form of asthma and mood disorders. Gene expression commonly regulated in PTSD and severe asthma, included ORMDL3 a gene known to be associated with asthma risk and STX8, which is involved in TrkA signaling. We also identified several pathways commonly regulated to both MDD and severe asthma. This study reveals gene and pathway regulation that potentially drives the comorbidity between severe asthma, PTSD, and MDD and may serve as foci for future research aimed at gaining a better understanding of both the relationship between asthma and PTSD, and the pathophysiology of the individual disorders.
Collapse
Affiliation(s)
- Sandor Haas-Neill
- The Brain Body Institute, St. Joseph’s Hospital, McMaster University, Hamilton, Ontario, Canada
| | - Anna Dvorkin-Gheva
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Paul Forsythe
- Alberta Respiratory Centre, Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
Amici F, Röder S, Kiess W, Borte M, Zenclussen AC, Widdig A, Herberth G. Maternal stress, child behavior and the promotive role of older siblings. BMC Public Health 2022; 22:863. [PMID: 35488325 PMCID: PMC9055772 DOI: 10.1186/s12889-022-13261-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/19/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND In the first years of their lives, children develop the cognitive, social and emotional skills that will provide the foundations for their lifelong health and achievements. To increase their life prospects and reduce the long-term effects of early aversive conditions, it is therefore crucial to understand the risk factors that negatively affect child development and the factors that are instead beneficial. In this study, we tested (i) the effects of different social and environmental stressors on maternal stress levels, (ii) the dynamic relationship between maternal stress and child behavior problems during development, and (iii) the potential promotive (i.e. main) or protective (i.e. buffering) effect of siblings on child behavior problems during development. METHODS We used longitudinal data from 373 mother-child pairs (188 daughters, 185 sons) from pregnancy until 10 years of age. We assessed maternal stress and child behavior problems (internalizing and externalizing) with validated questionnaires, and then used linear mixed models, generalized linear mixed models and longitudinal cross-lagged models to analyze the data. RESULTS Our results showed that higher maternal stress levels were predicted by socio-environmental stressors (i.e. the lack of sufficient social areas in the neighborhood). Moreover, prenatal maternal stress reliably predicted the occurrence of behavior problems during childhood. Finally, the presence of older siblings had a promotive function, by reducing the likelihood that children developed externalizing problems. CONCLUSIONS Overall, our results confirm the negative effects that maternal stress during pregnancy may have on the offspring, and suggest an important main effect of older siblings in promoting a positive child development.
Collapse
Affiliation(s)
- Federica Amici
- Department of Comparative Cultural Psychology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103, Leipzig, Germany.
- Behavioural Ecology Research Group, Institute of Biology, University of Leipzig, 04103, Leipzig, Germany.
| | - Stefan Röder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Wieland Kiess
- Department of Women and Child Health, Center of Paediatric Research (CPL), Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
- Leipzig Research Centre for Civilization Diseases (LIFE), Leipzig University, Leipzig, Germany
| | - Michael Borte
- Children's Hospital, Municipal Hospital "St. Georg", Academic Teaching Hospital of the University of Leipzig, 04129, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Anja Widdig
- Behavioural Ecology Research Group, Institute of Biology, University of Leipzig, 04103, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103, Leipzig, Germany
- Department of Human Behaviour, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, 04103, Leipzig, Germany
| | - Gunda Herberth
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| |
Collapse
|
11
|
Gautam Y, Johansson E, Mersha TB. Multi-Omics Profiling Approach to Asthma: An Evolving Paradigm. J Pers Med 2022; 12:jpm12010066. [PMID: 35055381 PMCID: PMC8778153 DOI: 10.3390/jpm12010066] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Asthma is a complex multifactorial and heterogeneous respiratory disease. Although genetics is a strong risk factor of asthma, external and internal exposures and their interactions with genetic factors also play important roles in the pathophysiology of asthma. Over the past decades, the application of high-throughput omics approaches has emerged and been applied to the field of asthma research for screening biomarkers such as genes, transcript, proteins, and metabolites in an unbiased fashion. Leveraging large-scale studies representative of diverse population-based omics data and integrating with clinical data has led to better profiling of asthma risk. Yet, to date, no omic-driven endotypes have been translated into clinical practice and management of asthma. In this article, we provide an overview of the current status of omics studies of asthma, namely, genomics, transcriptomics, epigenomics, proteomics, exposomics, and metabolomics. The current development of the multi-omics integrations of asthma is also briefly discussed. Biomarker discovery following multi-omics profiling could be challenging but useful for better disease phenotyping and endotyping that can translate into advances in asthma management and clinical care, ultimately leading to successful precision medicine approaches.
Collapse
|
12
|
Brew BK, Lundholm C, Caffrey Osvald E, Chambers G, Öberg S, Fang F, Almqvist C. Early-Life Adversity Due to Bereavement and Inflammatory Diseases in the Next Generation: A Population Study in Transgenerational Stress Exposure. Am J Epidemiol 2022; 191:38-48. [PMID: 34550338 PMCID: PMC8751780 DOI: 10.1093/aje/kwab236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/25/2021] [Accepted: 09/17/2021] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence suggests that trauma experienced in childhood has negative transgenerational implications for offspring mental and physical health. We aimed to investigate whether early-life adversity experienced as bereavement is associated with chronic inflammatory health in offspring. The study population included 3 generations of Swedish families with a base population of 453,516 children (generation 3) born in 2001–2012. Exposure was defined as the middle generation’s (generation 2) experiencing bereavement in childhood due to the death of a parent (generation 1). Outcomes in generation 3 included 2 diagnoses of inflammatory diseases, including asthma, allergic diseases, eczema, and autoimmune diseases. Survival analysis was used to identify causal pathways, including investigation of mediation by generation 2 mood disorders and socioeconomic status (SES). We found that early-life bereavement experienced by women was associated with early-onset offspring asthma (hazard ratio = 1.15, 95% confidence interval: 1.08, 1.23); mediation analysis revealed that 28%–33% of the association may be mediated by SES and 9%–20% by mood disorders. Early-life bereavement experienced by men was associated with autoimmune diseases in offspring (hazard ratio = 1.31, 95% confidence interval: 1.06, 1.62), with no evidence of mediation. In conclusion, adversity experienced early in life may contribute to an increased risk of inflammatory diseases which is partly mediated by mood disorders and SES.
Collapse
Affiliation(s)
- Bronwyn K Brew
- Correspondence to Dr. Bronwyn K. Brew, Department of Medical Epidemiology and Biostatistics, Karolinska Institute, 12a Nobels vag, Solna, 171 77, Stockholm, Sweden (e-mail: )
| | | | | | | | | | | | | |
Collapse
|
13
|
Ross MK, Romero T, Szilagyi PG. Adverse Childhood Experiences and Association With Pediatric Asthma Severity in the 2016-2017 National Survey of Children's Health. Acad Pediatr 2021; 21:1025-1030. [PMID: 33940207 PMCID: PMC8947844 DOI: 10.1016/j.acap.2021.04.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Prior studies have found that adverse childhood experiences (ACEs) are associated with asthma prevalence and onset, presumably related to stress and increased inflammation. We hypothesized that ACEs may be associated with asthma severity as well. We studied the 2016-2017 US National Survey of Children's Health dataset to explore the relationship between ACEs and pediatric asthma severity. METHODS We analyzed children ages 0 to 17 years old who had current caregiver-reported asthma diagnosed by a healthcare provider. We reported descriptive characteristics using chi-square analysis of variance (ANOVA) and used multivariable regression analysis to assess the relationship of cumulative and individual ACEs with asthma severity. Survey sampling weights and SAS survey procedures were implemented to produce nationally representative results. RESULTS Our analysis included 3691 children, representing a population of 5,465,926. Unadjusted analysis demonstrated that ACEs - particularly household economic hardship, parent/guardian served time in jail, witnessed household violence, or victim/witness of neighborhood violence - were each associated with higher odds of moderate/severe caregiver-reported asthma. After controlling for confounders possibly associated with both exposure (ACEs) and outcome (asthma severity), children who witnessed parent/adult violence had higher adjusted odds of caregiver-reported moderate/severe asthma. (1.67, confidence interval 1.05-2.64, P = .03) CONCLUSIONS: Intrafamilial witnessed household violence is significantly associated with caregiver-reported moderate/severe asthma.
Collapse
Affiliation(s)
- Mindy K. Ross
- University of California Los Angeles, Department of Pediatrics, Pediatric Pulmonology and Sleep Medicine, Los Angeles, Calif
| | - Tahmineh Romero
- University of California Los Angeles, Department of Medicine, Statistical Core, Los Angeles, Calif
| | - Peter G Szilagyi
- University of California Los Angeles, Department of Pediatrics, General Pediatrics, Los Angeles, Calif
| |
Collapse
|
14
|
Exposure to Stress and Air Pollution from Bushfires during Pregnancy: Could Epigenetic Changes Explain Effects on the Offspring? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147465. [PMID: 34299914 PMCID: PMC8305161 DOI: 10.3390/ijerph18147465] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022]
Abstract
Due to climate change, bushfires are becoming a more frequent and more severe phenomenon which contributes to poor health effects associated with air pollution. In pregnancy, environmental exposures can have lifelong consequences for the fetus, but little is known about these consequences in the context of bushfire smoke exposure. In this review we summarise the current knowledge in this area, and propose a potential mechanism linking bushfire smoke exposure in utero to poor perinatal and respiratory outcomes in the offspring. Bushfire smoke exposure is associated with poor pregnancy outcomes including reduced birth weight and an increased risk of prematurity. Some publications have outlined the adverse health effects on young children, particularly in relation to emergency department presentations and hospital admissions for respiratory problems, but there are no studies in children who were exposed to bushfire smoke in utero. Prenatal stress is likely to occur as a result of catastrophic bushfire events, and stress is known to be associated with poor perinatal and respiratory outcomes. Changes to DNA methylation are potential epigenetic mechanisms linking both smoke particulate exposure and prenatal stress to poor childhood respiratory health outcomes. More research is needed in large pregnancy cohorts exposed to bushfire events to explore this further, and to design appropriate mitigation interventions, in this area of global public health importance.
Collapse
|
15
|
Wan MW, Janta-Lipinski M, Osam CS. Childhood Allergies: The Role of Maternal Depression and Anxiety, and Family Strain. CHILDREN-BASEL 2021; 8:children8030185. [PMID: 33804405 PMCID: PMC7999446 DOI: 10.3390/children8030185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022]
Abstract
Maternal mental disorder and a negative family emotional climate are a great source of stress for many children, yet their role in the childhood development or expression of asthma and allergies remains poorly understood, particularly beyond the first 1–2 years of life. The current study tested whether childhood allergy onset and symptomatology would be predicted by (1) perinatal and any time exposure to maternal depression or anxiety and (2) current family emotional strain (whole family, mother-child). UK mothers of children aged 2–12 years (N = 328) living with them completed an online survey of measures. Children exposed to maternal depression were almost twice as likely to be diagnosed and almost five times as likely to screen positive for an allergic disorder. Perinatal depression was linked to childhood allergies, but more moderately. Any anxiety exposure, and not specific to the perinatal period, predicted allergy status. Family emotional strain contributed independently to variance in concurrent child allergic symptomatology. All results were independent of potential confounders and current mental distress. The findings highlight the importance of maternal mental health and family function in the child’s neuro-immune development, and that these factors need to be addressed in the treatment of childhood allergic disorders.
Collapse
Affiliation(s)
- Ming Wai Wan
- Perinatal Mental Health and Parenting Research Unit (PRIME-RU), School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Centre for Women’s Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Correspondence:
| | - Molly Janta-Lipinski
- Perinatal Mental Health and Parenting Research Unit (PRIME-RU), School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Cemre Su Osam
- Centre for Women’s Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| |
Collapse
|
16
|
Yamamoto-Hanada K, Pak K, Saito-Abe M, Sato M, Ohya Y. Better maternal quality of life in pregnancy yields better offspring respiratory outcomes: A birth cohort. Ann Allergy Asthma Immunol 2021; 126:713-721.e1. [PMID: 33639261 DOI: 10.1016/j.anai.2021.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/28/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND It is suggested that maternal mental health during pregnancy may affect offspring immune and respiratory features, based on the developmental origins of health and disease hypothesis. OBJECTIVE To evaluate whether maternal quality of life (QoL) and depression during pregnancy leads to wheezing, asthma, and food allergy of the offspring at 3 years of age. METHODS We conducted a nationwide, multicenter, prospective birth cohort study, Japan Environment and Children's Study. All variables were collected from questionnaires. Health-related QoL was measured using the Medical Outcomes Survey Short Form-8 questionnaire with a physical component summary and a mental component summary score. We conducted logistic regression analyses to evaluate the associations of offspring's wheezing, asthma, and food allergy with maternal QoL and depression. RESULTS There were 72,685 participants with no missing variables. Maternal physical component summary scores of the Medical Outcomes Survey Short Form-8 questionnaire were negatively associated with offspring's asthma (adjusted odds ratio [aOR], 0.99; 95% confidence interval [CI], 0.99-1.00), current wheezing (aOR, 0.99; 95% CI, 0.99-0.99), and food allergy diagnoses (aOR, 0.99; 95% CI, 0.98-0.99) in children. Offspring's wheezing and asthma were also associated with maternal depression and anxiety during pregnancy. CONCLUSION Poor maternal prenatal QoL increased the risk of wheezing, asthma, and food allergy in offspring. In addition, maternal depression and anxiety increased the risk of offspring's wheezing, asthma, and food allergy.
Collapse
Affiliation(s)
- Kiwako Yamamoto-Hanada
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan; Medical Support Center for the Japan Environment and Children's Study, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Kyongsun Pak
- Division of Biostatistics, Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Mayako Saito-Abe
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan; Medical Support Center for the Japan Environment and Children's Study, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Miori Sato
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan; Medical Support Center for the Japan Environment and Children's Study, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan; Medical Support Center for the Japan Environment and Children's Study, National Research Institute for Child Health and Development, Tokyo, Japan
| | | |
Collapse
|
17
|
van Meel ER, Saharan G, Jaddoe VW, de Jongste JC, Reiss IK, Tiemeier H, El Marroun H, Duijts L. Parental psychological distress during pregnancy and the risk of childhood lower lung function and asthma: a population-based prospective cohort study. Thorax 2020; 75:1074-1081. [PMID: 33046570 PMCID: PMC7677473 DOI: 10.1136/thoraxjnl-2019-214099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 11/16/2022]
Abstract
Background Although maternal psychological distress during pregnancy is associated with increased risks of respiratory morbidity in preschool children, it is unknown whether this association persists into later childhood. Objective To examine the association between parental psychological distress during pregnancy and lung function and asthma in children of school age. Methods This study of 4231 children was embedded in a population-based prospective cohort. Parental psychological distress was assessed by the Brief Symptom Inventory during and 3 years after pregnancy, and in mothers also at 2 and 6 months after pregnancy. At age 10 years, lung function was obtained by spirometry and asthma by questionnaire. Results The prevalence of asthma was 5.9%. Maternal overall psychological distress during pregnancy was associated with a lower forced vital capacity (FVC) (z-score difference −0.10 (95% CI −0.20 to –0.01) per 1-unit increase), maternal depressive symptoms during pregnancy with a lower forced expiratory volume in the first second (FEV1) and FVC (−0.13 (95% CI −0.24 to –0.01) and −0.13 (95% CI −0.24 to –0.02) when using clinical cut-offs) in their children. All maternal psychological distress measures during pregnancy were associated with an increased risk of asthma (range OR: 1.46 (95% CI 1.12 to 1.90) to 1.91 (95% CI 1.26 to 2.91)). Additional adjustment for paternal psychological distress during pregnancy and parental psychological distress after pregnancy did not materially change the associations. Paternal psychological distress during pregnancy was not associated with childhood respiratory morbidity. Conclusion Maternal, but not paternal, psychological distress during pregnancy is associated with an increased risk of asthma and partly lower lung function in children. This suggests intrauterine programming for the risk of later-life respiratory disease.
Collapse
Affiliation(s)
- Evelien R van Meel
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gautam Saharan
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vincent Wv Jaddoe
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Irwin Km Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Social and Behavioural Science, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Hanan El Marroun
- Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Psychology, Education and Child Studies, Erasmus Universiteit Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus Medical Center, Rotterdam, The Netherlands .,Department of Pediatrics, Division of Neonatology, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| |
Collapse
|
18
|
Dong E, Pandey SC. Prenatal stress induced chromatin remodeling and risk of psychopathology in adulthood. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 156:185-215. [PMID: 33461663 PMCID: PMC7864549 DOI: 10.1016/bs.irn.2020.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
New insights into the pathophysiology of psychiatric disorders suggest the existence of a complex interplay between genetics and environment. This notion is supported by evidence suggesting that exposure to stress during pregnancy exerts profound effects on the neurodevelopment and behavior of the offspring and predisposes them to psychiatric disorders later in life. Accumulated evidence suggests that vulnerability to psychiatric disorders may result from permanent negative effects of long-term changes in synaptic plasticity due to altered epigenetic mechanisms (histone modifications and DNA methylation) that lead to condensed chromatin architecture, thereby decreasing the expression of candidate genes during early brain development. In this chapter, we have summarized the literature of clinical studies on psychiatric disorders induced by maternal stress during pregnancy. We also discussed the epigenetic alterations of gene regulations induced by prenatal stress. Because the clinical manifestations of psychiatric disorders are complex, it is obvious that the biological progression of these diseases cannot be studied only in postmortem brains of patients and the use of animal models is required. Therefore, in this chapter, we have introduced a well-established mouse model of prenatal stress (PRS) generated in restrained pregnant dams. The behavioral phenotypes of the offspring (PRS mice) born to the stressed dam and underlying epigenetic changes in key molecules related to synaptic activity were described and highlighted. PRS mice may serve as a useful model for investigating the pathogenesis of psychiatric disorders and may be a useful tool for screening for the potential compounds that may normalize aberrant epigenetic mechanisms induced by prenatal stress.
Collapse
Affiliation(s)
- Erbo Dong
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
19
|
Landeo-Gutierrez J, Forno E, Miller GE, Celedón JC. Exposure to Violence, Psychosocial Stress, and Asthma. Am J Respir Crit Care Med 2020; 201:917-922. [PMID: 31801032 PMCID: PMC7159436 DOI: 10.1164/rccm.201905-1073pp] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/04/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jeremy Landeo-Gutierrez
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Erick Forno
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Gregory E. Miller
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, Illinois
| | - Juan C. Celedón
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| |
Collapse
|
20
|
Transcriptome signatures from discordant sibling pairs reveal changes in peripheral blood immune cell composition in Autism Spectrum Disorder. Transl Psychiatry 2020; 10:106. [PMID: 32291385 PMCID: PMC7156413 DOI: 10.1038/s41398-020-0778-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022] Open
Abstract
Notwithstanding several research efforts in the past years, robust and replicable molecular signatures for autism spectrum disorders from peripheral blood remain elusive. The available literature on blood transcriptome in ASD suggests that through accurate experimental design it is possible to extract important information on the disease pathophysiology at the peripheral level. Here we exploit the availability of a resource for molecular biomarkers in ASD, the Italian Autism Network (ITAN) collection, for the investigation of transcriptomic signatures in ASD based on a discordant sibling pair design. Whole blood samples from 75 discordant sibling pairs selected from the ITAN network where submitted to RNASeq analysis and data analyzed by complementary approaches. Overall, differences in gene expression between affected and unaffected siblings were small. In order to assess the contribution of differences in the relative proportion of blood cells between discordant siblings, we have applied two different cell deconvolution algorithms, showing that the observed molecular signatures mainly reflect changes in peripheral blood immune cell composition, in particular NK cells. The results obtained by the cell deconvolution approach are supported by the analysis performed by WGCNA. Our report describes the largest differential gene expression profiling in peripheral blood of ASD subjects and controls conducted by RNASeq. The observed signatures are consistent with the hypothesis of immune alterations in autism and an increased risk of developing autism in subjects exposed to prenatal infections or stress. Our study also points to a potential role of NMUR1, HMGB3, and PTPRN2 in ASD.
Collapse
|
21
|
Jafari Z, Kolb BE, Mohajerani MH. Life-Course Contribution of Prenatal Stress in Regulating the Neural Modulation Network Underlying the Prepulse Inhibition of the Acoustic Startle Reflex in Male Alzheimer's Disease Mice. Cereb Cortex 2020; 30:311-325. [PMID: 31070710 PMCID: PMC7029700 DOI: 10.1093/cercor/bhz089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The prepulse inhibition (PPI) of the acoustic startle reflex (ASR), as an index of sensorimotor gating, is one of the most extensively used paradigms in the field of neuropsychiatric disorders. Few studies have examined how prenatal stress (PS) regulates the sensorimotor gating during the lifespan and how PS modifies the development of amyloid-beta (Aβ) pathology in brain areas underlying the PPI formation. We followed alternations in corticosterone levels, learning and memory, and the PPI of the ASR measures in APPNL-G-F/NL-G-F offspring of dams exposed to gestational noise stress. In-depth quantifications of the Aβ plaque accumulation were also performed at 6 months. The results indicated an age-dependent deterioration of sensorimotor gating, long-lasting PS-induced abnormalities in PPI magnitudes, as well as deficits in spatial memory. The PS also resulted in a higher Aβ aggregation predominantly in brain areas associated with the PPI modulation network. The findings suggest the contribution of a PS-induced hypothalamic-pituitary-adrenal (HPA) axis hyperactivity in regulating the PPI modulation substrates leading to the abnormal development of the neural protection system in response to disruptive stimuli. The long-lasting HPA axis dysregulation appears to be the major underlying mechanism in precipitating the Aβ deposition, especially in brain areas contributed to the PPI modulation network.
Collapse
Affiliation(s)
- Zahra Jafari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
- Department of Basic Sciences in Rehabilitation, School of Rehabilitation Sciences, Iran University of Medical Science, Tehran, Iran
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| |
Collapse
|
22
|
Klös M, Thürmann L, Bauer M, Heinrich J, Standl M, von Berg A, Schaaf B, Weichenhan D, Mücke O, Plass C, Röder S, Herberth G, Sack U, Borte M, Stangl GI, Eils R, Trump S, Lehmann I. Longitudinal trends of serum IgE and IL5RA expression throughout childhood are associated with asthma but not with persistent wheeze. Allergy 2019; 74:2002-2006. [PMID: 31012487 DOI: 10.1111/all.13837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Matthias Klös
- Molecular Epidemiology Unit Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, associated partner of the German Center for Lung Research (DZL) Berlin Germany
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Loreen Thürmann
- Molecular Epidemiology Unit Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, associated partner of the German Center for Lung Research (DZL) Berlin Germany
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Mario Bauer
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Joachim Heinrich
- Institute and Clinic for Occupational, Social and Environmental Medicine University HospitalLMU Munich Germany
- Institute of Epidemiology Helmholtz Zentrum München ‐ German ResearchCenter for Environmental Health Neuherberg Germany
| | - Marie Standl
- Institute of Epidemiology Helmholtz Zentrum München ‐ German ResearchCenter for Environmental Health Neuherberg Germany
| | - Andrea von Berg
- Department of Pediatrics Research Institute Marien‐Hospital Wesel Wesel Germany
| | - Beate Schaaf
- Pediatric Outpatient Department Bad Honnef Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics German Cancer Research Center (DKFZ)Heidelberg Germany
| | - Oliver Mücke
- Division of Cancer Epigenomics German Cancer Research Center (DKFZ)Heidelberg Germany
| | - Christoph Plass
- Division of Cancer Epigenomics German Cancer Research Center (DKFZ)Heidelberg Germany
| | - Stefan Röder
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Gunda Herberth
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Ulrich Sack
- Medical Faculty Institute for Clinical Immunology University of Leipzig Leipzig Germany
| | - Michael Borte
- Children's HospitalMunicipal Hospital “St. Georg” Leipzig Germany
| | - Gabriele I. Stangl
- Institute of Agricultural and Nutritional Sciences Martin Luther University Halle‐Wittenberg Halle Germany
| | - Roland Eils
- Center for Digital Health Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, associated partner of the German Center for Lung Research (DZL) Berlin Germany
- Heidelberg University HospitalHealth Data Science Unit Heidelberg Germany
| | - Saskia Trump
- Molecular Epidemiology Unit Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, associated partner of the German Center for Lung Research (DZL) Berlin Germany
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| | - Irina Lehmann
- Molecular Epidemiology Unit Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, associated partner of the German Center for Lung Research (DZL) Berlin Germany
- Department of Environmental Immunology Helmholtz Centre for EnvironmentalResearch – UFZ Leipzig Germany
| |
Collapse
|
23
|
Rusconi F, Gagliardi L. Pregnancy Complications and Wheezing and Asthma in Childhood. Am J Respir Crit Care Med 2019; 197:580-588. [PMID: 29064265 DOI: 10.1164/rccm.201704-0744pp] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Franca Rusconi
- 1 Epidemiology Unit, Anna Meyer Children's University Hospital, Florence, Italy; and
| | - Luigi Gagliardi
- 2 Pediatrics and Neonatology Division, Versilia Hospital, Azienda Toscana Nord Ovest, Pisa, Italy
| |
Collapse
|
24
|
Richardson TG, Richmond RC, North TL, Hemani G, Davey Smith G, Sharp GC, Relton CL. An integrative approach to detect epigenetic mechanisms that putatively mediate the influence of lifestyle exposures on disease susceptibility. Int J Epidemiol 2019; 48:887-898. [PMID: 31257439 PMCID: PMC6659375 DOI: 10.1093/ije/dyz119] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND There is mounting evidence that our environment and lifestyle has an impact on epigenetic regulatory mechanisms, such as DNA methylation. It has been suggested that these molecular processes may mediate the effect of risk factors on disease susceptibility, although evidence in this regard has been challenging to uncover. Using genetic variants as surrogate variables, we have used two-sample Mendelian randomization (2SMR) to investigate the potential implications of putative changes to DNA methylation levels on disease susceptibility. METHODS To illustrate our approach, we identified 412 CpG sites where DNA methylation was associated with prenatal smoking. We then applied 2SMR to investigate potential downstream effects of these putative changes on 643 complex traits using findings from large-scale genome-wide association studies. To strengthen evidence of mediatory mechanisms, we used multiple-trait colocalization to assess whether DNA methylation, nearby gene expression and complex trait variation were all influenced by the same causal genetic variant. RESULTS We identified 22 associations that survived multiple testing (P < 1.89 × 10-7). In-depth follow-up analyses of particular note suggested that the associations between DNA methylation at the ASPSCR1 and REST/POL2RB gene regions, both linked with reduced lung function, may be mediated by changes in gene expression. We validated associations between DNA methylation and traits using independent samples from different stages across the life course. CONCLUSION Our approach should prove valuable in prioritizing CpG sites that may mediate the effect of causal risk factors on disease. In-depth evaluations of findings are necessary to robustly disentangle causality from alternative explanations such as horizontal pleiotropy.
Collapse
Affiliation(s)
- Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Teri-Louise North
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| |
Collapse
|
25
|
Kim HB. Modifiable prenatal environmental factors for the prevention of childhood asthma. ALLERGY ASTHMA & RESPIRATORY DISEASE 2019; 7:179. [DOI: 10.4168/aard.2019.7.4.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/14/2019] [Indexed: 09/01/2023]
Affiliation(s)
- Hyo-Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, Korea
| |
Collapse
|
26
|
Maternal psychological distress during pregnancy and childhood health outcomes: a narrative review. J Dev Orig Health Dis 2018; 10:274-285. [PMID: 30378522 DOI: 10.1017/s2040174418000557] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal psychological distress is common in pregnancy and may influence the risk of adverse outcomes in children. Psychological distress may cause a suboptimal intrauterine environment leading to growth and developmental adaptations of the fetus and child. In this narrative review, we examined the influence of maternal psychological distress during pregnancy on fetal outcomes and child cardiometabolic, respiratory, atopic and neurodevelopment-related health outcomes. We discussed these findings from an epidemiological and life course perspective and provided recommendations for future studies. The literature in the field of maternal psychological distress and child health outcomes is extensive and shows that exposure to stress during pregnancy is associated with multiple adverse child health outcomes. Because maternal psychological distress is an important and potential modifiable factor during pregnancy, it should be a target for prevention strategies in order to optimize fetal and child health. Future studies should use innovative designs and strategies in order to address the issue of causality.
Collapse
|
27
|
Early maternal perceived stress and children's BMI: longitudinal impact and influencing factors. BMC Public Health 2018; 18:1211. [PMID: 30376822 PMCID: PMC6208039 DOI: 10.1186/s12889-018-6110-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/11/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Maternal perceived stress has been discussed to contribute to the development of childhood overweight. Our aim was to investigate the longitudinal relationship of early maternal perceived stress and BMI z-scores in preschool children (≤ five years). METHODS A longitudinal analysis was conducted in 498 mother-child pairs of the German prospective birth cohort LINA with information on maternal perceived stress during pregnancy, one and two years after birth. BMI z-scores were based on annual measurements of children's weight/height and calculated based on WHO reference data. General estimation equations were applied to evaluate the impact of maternal stress on children's longitudinal BMI z-scores. Potential stressors contributing to the perceived stress of the mother were assessed by linear regression models. Using mediation analyses we evaluated the relationship between stressors, maternal perceived stress, and children's BMI z-score development. RESULTS Postnatal maternal stress during the first year after birth had a positive longitudinal relationship with children's BMI z-scores up to the age of five years. Gender-stratified analyses revealed that only girls showed this positive association while boy's BMI z-scores were unaffected by maternal stress. We identified three neighborhood strains and two socio-demographic factors, which contributed to the maternal perceived stress level. Stressors themselves did not directly affect girl's BMI z-scores but rather mediated their effect through the perceived stress level. CONCLUSIONS While different stressors contribute to maternal stress, the perceived stress level - rather than the stressors themselves - is strongly positively associated with BMI z-score development in girls.
Collapse
|
28
|
Brunst KJ, Tignor N, Just A, Liu Z, Lin X, Hacker MR, Bosquet Enlow M, Wright RO, Wang P, Baccarelli AA, Wright RJ. Cumulative lifetime maternal stress and epigenome-wide placental DNA methylation in the PRISM cohort. Epigenetics 2018; 13:665-681. [PMID: 30001177 DOI: 10.1080/15592294.2018.1497387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Evolving evidence links maternal stress exposure to changes in placental DNA methylation of specific genes regulating placental function that may have implications for the programming of a host of chronic disorders. Few studies have implemented an epigenome-wide approach. Using the Infinium HumanMethylation450 BeadChip (450K), we investigated epigenome-wide placental DNA methylation in relation to maternal experiences of traumatic and non-traumatic stressors over her lifetime assessed using the Life Stressor Checklist-Revised (LSC-R) survey (n = 207). We found differential DNA methylation at epigenome-wide statistical significance (FDR = 0.05) for 112 CpGs. Additionally, we observed three clusters that exhibited differential methylation in response to high maternal lifetime stress. Enrichment analyses, conducted at an FDR = 0.20, revealed lysine degradation to be the most significant pathway associated with maternal lifetimes stress exposure. Targeted enrichment analyses of the three largest clusters of probes, identified using the gap statistic, were enriched for genes associated with endocytosis (i.e., SMAP1, ANKFY1), tight junctions (i.e., EPB41L4B), and metabolic pathways (i.e., INPP5E, EEF1B2). These pathways, also identified in the top 10 KEGG pathways associated with maternal lifetime stress exposure, play important roles in multiple physiological functions necessary for proper fetal development. Further, two genes were identified to exhibit multiple probes associated with maternal lifetime stress (i.e., ANKFY1, TM6SF1). The methylation status of the probes belonging to each cluster and/or genes exhibiting multiple hits, may play a role in the pathogenesis of adverse health outcomes in children born to mothers with increased lifetime stress exposure.
Collapse
Affiliation(s)
- Kelly J Brunst
- a Department of Environmental Health , University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Nicole Tignor
- b Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences , Icahn School of Medicine at Mount Sinai One Gustave L. Levy Place , New York , NY , USA
| | - Allan Just
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Zhonghua Liu
- d Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Xihong Lin
- d Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Michele R Hacker
- e Department of Obstetrics and Gynecology , Beth Israel Deaconess Medical Center , Boston , MA , USA.,f Department of Obstetrics , Gynecology and Reproductive Biology, Harvard Medical School , Boston , MA , USA
| | - Michelle Bosquet Enlow
- g Department of Psychiatry, Program for Behavioral Science, Boston Children's Hospital and Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| | - Robert O Wright
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Pei Wang
- b Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences , Icahn School of Medicine at Mount Sinai One Gustave L. Levy Place , New York , NY , USA
| | - Andrea A Baccarelli
- h Department of Environmental Health Sciences , Mailman School of Public Health, Columbia University , New York , NY , USA
| | - Rosalind J Wright
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA.,i Department of Pediatrics , Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai , New York , NY , USA
| |
Collapse
|
29
|
Radhakrishnan D, Shariff SZ, To T. The influence of prenatal mental health service use on the incidence of childhood asthma: a population-based cohort study. J Asthma 2018; 56:395-403. [PMID: 29693465 DOI: 10.1080/02770903.2018.1466313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES We aimed to determine whether maternal mental health service use during pregnancy, a potential proxy measure of prenatal maternal stress, is associated with the development of asthma in a large population-based sample of children. We hypothesized that children born to mothers with mental health service use during pregnancy would have a higher incidence of childhood asthma. STUDY DESIGN Health administrative data from Ontario, Canada (population >13 million) was used to identify pairs of mothers linked with their children born between April 1, 2001 to March 31, 2002. Descriptive statistics were used to compare the cumulative incidence of asthma by age 12 years in children whose mothers did or did not have prenatal mental health service use. Multivariable logistic regression was used to estimate the association between prenatal maternal mental health service use and childhood asthma incidence, after adjusting for the child's sex, residency (rural vs. urban), socioeconomic status, comorbid health conditions, low birthweight, and maternal history of asthma. RESULTS In a population-based sample of 122,333 children, those born to mothers with mental health service use during pregnancy had increased odds of developing asthma (odds ratio: 1.16, 95% confidence intervals: 1.12, 1.20, p < 0.001). CONCLUSIONS Prenatal maternal mental health service use is an independent risk factor for the development of asthma in childhood. This supports growing evidence for the importance of in utero exposure to maternal stress factors in asthma pathogenesis. This study highlights a potential strategy for the primary prevention of childhood asthma, namely improved recognition and management of mental health issues and stress in pregnant mothers.
Collapse
Affiliation(s)
- Dhenuka Radhakrishnan
- a Department of Pediatrics , Children's Hospital of Eastern Ontario , Ottawa , Ontario , Canada.,b Department of Pediatrics , University of Ottawa , Ottawa , Ontario , Canada.,c Department of Pediatrics , Institute for Clinical Evaluative Sciences , Ontario , Canada
| | - Salimah Z Shariff
- c Department of Pediatrics , Institute for Clinical Evaluative Sciences , Ontario , Canada
| | - Teresa To
- c Department of Pediatrics , Institute for Clinical Evaluative Sciences , Ontario , Canada.,d Department of Pediatrics , Hospital for Sick Children, Child Health Evaluative Sciences , Toronto , Ontario , Canada.,e Department of Pediatrics , Dalla Lana School of Public Health, University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|
30
|
Magnus MC, Wright RJ, Røysamb E, Parr CL, Karlstad Ø, Page CM, Nafstad P, Håberg SE, London SJ, Nystad W. Association of Maternal Psychosocial Stress With Increased Risk of Asthma Development in Offspring. Am J Epidemiol 2018; 187:1199-1209. [PMID: 29244063 PMCID: PMC5982733 DOI: 10.1093/aje/kwx366] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
Prenatal maternal psychosocial stress might influence the development of childhood asthma. Evaluating paternal psychosocial stress and conducting a sibling comparison could provide further insight into the role of unmeasured confounding. We examined the associations of parental psychosocial stress during and after pregnancy with asthma at age 7 years in the Norwegian Mother and Child Cohort Study (n = 63,626; children born in 2000-2007). Measures of psychosocial stress included lifetime major depressive symptoms, current anxiety/depression symptoms, use of antidepressants, anxiolytics, and/or hypnotics, life satisfaction, relationship satisfaction, work stress, and social support. Childhood asthma was associated with maternal lifetime major depressive symptoms (adjusted relative risk (aRR) = 1.19, 95% confidence interval (CI): 1.09, 1.30), in addition to symptoms of anxiety/depression during pregnancy (aRR = 1.17, 95% CI: 1.06, 1.29) and 6 months after delivery (aRR = 1.17, 95% CI: 1.07, 1.28). Maternal negative life events during pregnancy (aRR = 1.10, 95% CI: 1.06, 1.13) and 6 months after delivery (aRR = 1.14, 95% CI: 1.11, 1.18) were also associated with asthma. These associations were not replicated when evaluated within sibling groups. There were no associations with paternal psychosocial stress. In conclusion, maternal anxiety/depression and negative life events were associated with offspring asthma, but this might be explained by unmeasured maternal background characteristics that remain stable across deliveries.
Collapse
Affiliation(s)
- Maria C Magnus
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Rosalind J Wright
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Espen Røysamb
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Psychology, Faculty of Social Sciences, University of Oslo, Oslo, Norway
| | - Christine L Parr
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Øystein Karlstad
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Christian M Page
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Per Nafstad
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Community Medicine, Medical Faculty, University of Oslo, Oslo, Norway
| | - Siri E Håberg
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Wenche Nystad
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
31
|
Junge KM, Leppert B, Jahreis S, Wissenbach DK, Feltens R, Grützmann K, Thürmann L, Bauer T, Ishaque N, Schick M, Bewerunge-Hudler M, Röder S, Bauer M, Schulz A, Borte M, Landgraf K, Körner A, Kiess W, von Bergen M, Stangl GI, Trump S, Eils R, Polte T, Lehmann I. MEST mediates the impact of prenatal bisphenol A exposure on long-term body weight development. Clin Epigenetics 2018; 10:58. [PMID: 29721103 PMCID: PMC5910578 DOI: 10.1186/s13148-018-0478-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 03/23/2018] [Indexed: 01/01/2023] Open
Abstract
Background Exposure to endocrine-disrupting chemicals can alter normal physiology and increase susceptibility to non-communicable diseases like obesity. Especially the prenatal and early postnatal period is highly vulnerable to adverse effects by environmental exposure, promoting developmental reprogramming by epigenetic alterations. To obtain a deeper insight into the role of prenatal bisphenol A (BPA) exposure in children's overweight development, we combine epidemiological data with experimental models and BPA-dependent DNA methylation changes. Methods BPA concentrations were measured in maternal urine samples of the LINA mother-child-study obtained during pregnancy (n = 552), and BPA-associated changes in cord blood DNA methylation were analyzed by Illumina Infinium HumanMethylation450 BeadChip arrays (n = 472). Methylation changes were verified by targeted MassARRAY analyses, assessed for their functional translation by qPCR and correlated with children's body mass index (BMI) z scores at the age of 1 and 6 years. Further, female BALB/c mice were exposed to BPA from 1 week before mating until delivery, and weight development of their pups was monitored (n ≥ 8/group). Additionally, human adipose-derived mesenchymal stem cells were treated with BPA during the adipocyte differentiation period and assessed for exposure-related epigenetic, transcriptional and morphological changes (n = 4). Results In prenatally BPA-exposed children two CpG sites with deviating cord blood DNA-methylation profiles were identified, among them a hypo-methylated CpG in the promoter of the obesity-associated mesoderm-specific transcript (MEST). A mediator analysis suggested that prenatal BPA exposure was connected to cord blood MEST promoter methylation and MEST expression as well as BMI z scores in early infancy. This effect could be confirmed in mice in which prenatal BPA exposure altered Mest promoter methylation and transcription with a concomitant increase in the body weight of the juvenile offspring. An experimental model of in vitro differentiated human mesenchymal stem cells also revealed an epigenetically induced MEST expression and enhanced adipogenesis following BPA exposure. Conclusions Our study provides evidence that MEST mediates the impact of prenatal BPA exposure on long-term body weight development in offspring by triggering adipocyte differentiation.
Collapse
Affiliation(s)
- Kristin M. Junge
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Beate Leppert
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Susanne Jahreis
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Dirk K. Wissenbach
- Department Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Institute of Forensic Medicine, University Hospital Jena, Jena, Germany
| | - Ralph Feltens
- Department Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Konrad Grützmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT) Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Loreen Thürmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Berlin Institute of Health and Charité-Universitätsmedizin Berlin, Center for Digital Health, Berlin, Germany
| | - Tobias Bauer
- German Cancer Research Center (DKFZ), Division of Theoretical Bioinformatics, Heidelberg, Germany
| | - Naveed Ishaque
- German Cancer Research Center (DKFZ), Division of Theoretical Bioinformatics, Heidelberg, Germany
- Institute of Agriculture and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Matthias Schick
- German Cancer Research Center (DKFZ), Genomics and Proteomics Core Facility, Heidelberg, Germany
| | | | - Stefan Röder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Mario Bauer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Angela Schulz
- Medical Faculty, Rudolf-Schönheimer-Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Michael Borte
- Children’s Hospital, Municipal Hospital “St. Georg”, Leipzig, Germany
| | - Kathrin Landgraf
- LIFE-Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Hospital for Children and Adolescents-Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- LIFE-Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Hospital for Children and Adolescents-Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Wieland Kiess
- LIFE-Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Hospital for Children and Adolescents-Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Martin von Bergen
- Department Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Gabriele I. Stangl
- Institute of Agriculture and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena Leipzig, Germany
| | - Saskia Trump
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Roland Eils
- German Cancer Research Center (DKFZ), Heidelberg Center for Personalized Oncology, DKFZ-HIPO, Heidelberg, Germany
- Berlin Institute of Health and Charité-Universitätsmedizin Berlin, Center for Digital Health, Berlin, Germany
- Health Data Science Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Polte
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Irina Lehmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
- Unit for Molecular Epidemiology, Berlin Institute of Health (BIH) and Charitè - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
32
|
Miller RL, Lawrence J. Understanding Root Causes of Asthma. Perinatal Environmental Exposures and Epigenetic Regulation. Ann Am Thorac Soc 2018; 15:S103-S108. [PMID: 29676631 PMCID: PMC5946504 DOI: 10.1513/annalsats.201706-514mg] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/30/2017] [Indexed: 01/09/2023] Open
Abstract
A common explanation for the origins and rising prevalence of asthma is that they involve complex interactions between hereditary predispositions and environmental exposures that are incompletely understood. Yet, emerging evidence substantiates the paradigm that environmental exposures prenatally and during very early childhood induce epigenetic alterations that affect the expression of asthma genes and, thereby, asthma itself. Here, we review much of the key evidence supporting this paradigm. First, we describe evidence that the prenatal and early postnatal periods are key time windows of susceptibility to environmental exposures that may trigger asthma. Second, we explain how environmental epigenetic regulation may explain the immunopathology underlying asthma. Third, we outline specific evidence that environmental exposures induce epigenetic regulation, both from animal models and robust human epidemiological research. Finally, we review some emerging topics, including the importance of coexposures, population divergence, and how epigenetic regulation may change over time. Despite all the inherent complexity, great progress has been made toward understanding what we still consider reversible asthma risk factors. These, in time, may impact patient care.
Collapse
Affiliation(s)
- Rachel L. Miller
- Division of Pulmonary, Allergy and Critical Care of Medicine, Department of Medicine, and
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Jennifer Lawrence
- Division of Pulmonary, Allergy and Critical Care of Medicine, Department of Medicine, and
| |
Collapse
|
33
|
Rosa MJ, Lee A, Wright RJ. Evidence establishing a link between prenatal and early-life stress and asthma development. Curr Opin Allergy Clin Immunol 2018; 18:148-158. [PMID: 29369067 PMCID: PMC5835351 DOI: 10.1097/aci.0000000000000421] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to provide an update on our evolving understanding of the effects of stress in pregnancy and during early development on the onset of asthma-related phenotypes across childhood, adolescence, and into early adulthood. RECENT FINDINGS Accumulating evidence over the past 2 decades has established that prenatal and early-life psychological stress and stress correlates (e.g., maternal anxiety or depression) increase the risk for childhood respiratory disorders. Recent systematic reviews and meta-analyses including numerous prospective epidemiological and case-control studies substantiate a significant effect of prenatal stress and stress in early childhood on the development of wheeze, asthma, and other atopic-related disorders (eczema and allergic rhinitis), with many studies showing an exposure-response relationship. Offspring of both sexes are susceptible to perinatal stress, but effects differ. The impact of stress on child wheeze/asthma can also be modified by exposure timing. Moreover, coexposure to prenatal stress can enhance the effect of chemical stressors, such as prenatal traffic-related air pollution, on childhood respiratory disease risk. Understanding complex interactions among exposure dose, timing, child sex, and concurrent environmental exposures promises to more fully characterize stress effects and identify susceptible subgroups. Although the link between perinatal stress and childhood asthma-related phenotypes is now well established, pathways by which stress predisposes children to chronic respiratory disorders are not as well delineated. Mechanisms central to the pathophysiology of wheeze/asthma and lung growth and development overlap and involve a cascade of events that include disrupted immune, neuroendocrine, and autonomic function as well as oxidative stress. Altered homeostatic functioning of these integrated systems during development can enhance vulnerability to asthma and altered lung development. SUMMARY Mechanistic studies that more comprehensively assess biomarkers reflecting alterations across interrelated stress response systems and associated regulatory processes, in both pregnant women and young children, could be highly informative. Leveraging high-throughput systems-wide technologies to include epigenomics (e.g., DNA methylation, microRNAs), transcriptomics, and microbiomics as well as integrated multiomics are needed to advance this field of science. Understanding stress-induced physiological changes occurring during vulnerable life periods that contribute to chronic respiratory disease risk could lead to the development of preventive strategies and novel therapeutic interventions.
Collapse
Affiliation(s)
- Maria José Rosa
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison Lee
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
34
|
Flanigan C, Sheikh A, DunnGalvin A, Brew BK, Almqvist C, Nwaru BI. Prenatal maternal psychosocial stress and offspring's asthma and allergic disease: A systematic review and meta-analysis. Clin Exp Allergy 2018; 48:403-414. [PMID: 29331049 DOI: 10.1111/cea.13091] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/17/2017] [Accepted: 12/21/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Prenatal maternal stress may influence offspring's atopic risk through sustained cortisol secretion resulting from activation of the hypothalamic-pituitary axis (HPA), leading to Th2-biased cell differentiation in the foetus. We undertook a systematic review and meta-analysis investigating the relationship between prenatal maternal psychosocial stress and risk of asthma and allergy in the offspring. METHODS We searched 11 electronic databases from 1960 to 2016, searched the grey literature and contacted experts in the field. Type of stress indicator included mood disorders, anxiety, exposure to violence, bereavement and socio-economic problems occurring during pregnancy, both objectively and subjectively measured. We included all possible asthma and IgE-mediated allergy outcomes. We conducted random-effects meta-analyses to synthesize the data. RESULTS We identified 9779 papers of which 30 studies (enrolling >6 million participants) satisfied inclusion criteria. The quality of 25 studies was moderate, 4 were strong, and one was weak. Maternal exposure to any type of stressors was associated with an increased risk of offspring atopic eczema/dermatitis (OR 1.34, 95% CI 1.22-1.47), allergic rhinitis (OR 1.30, 95% CI 1.04-1.62), wheeze (OR 1.34, 95% CI 1.16-1.54) and asthma (OR 1.15, 95% CI 1.04-1.27). Exposure to anxiety and depression had strongest effect compared to other stressors. Exposure during the third trimester had the greatest impact compared to first and second trimesters. The increased risk was stronger for early-onset and persistent than for late-onset wheeze. Bereavement of a child (HR 1.28, 95% CI 1.10-1.48) or a spouse (HR 1.40, 95% CI 1.03-1.90) increased the risk of offspring asthma. CONCLUSIONS Exposure to prenatal maternal psychosocial stress was associated with increased risk, albeit modestly, of asthma and allergy in the offspring. The pronounced risk during the third trimester may represent cumulative stress exposure throughout pregnancy rather than trimester-specific effect. Our findings may represent a causal effect or a result of inherent biases in studies, particularly residual confounding.
Collapse
Affiliation(s)
- C Flanigan
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
| | - A Sheikh
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
| | - A DunnGalvin
- Department of Paediatrics and Child Health, Cork University Hospital, Cork City, Ireland.,University College Cork, Cork City, Ireland.,School of Applied Psychology, University College Cork, Cork City, Ireland
| | - B K Brew
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - C Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.,Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - B I Nwaru
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK.,Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,School of Health Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
35
|
Thürmann L, Grützmann K, Klös M, Bieg M, Winter M, Polte T, Bauer T, Schick M, Bewerunge-Hudler M, Röder S, Bauer M, Wissenbach DK, Sack U, Weichenhan D, Mücke O, Plass C, Borte M, von Bergen M, Lehmann I, Eils R, Trump S. Early-onset childhood atopic dermatitis is related to NLRP2 repression. J Allergy Clin Immunol 2017; 141:1482-1485.e16. [PMID: 29233739 DOI: 10.1016/j.jaci.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Loreen Thürmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Konrad Grützmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany; Center for Molecular Tumor Diagnostics at the National Center for Tumor Diseases, partner site Dresden, Dresden, Germany
| | - Matthias Klös
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Matthias Bieg
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Winter
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Tobias Polte
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Tobias Bauer
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Schick
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Bewerunge-Hudler
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Röder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Mario Bauer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Dirk K Wissenbach
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany; Institute of Forensic Medicine, University Hospital Jena, Jena, Germany
| | - Ulrich Sack
- Institute of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Dieter Weichenhan
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Mücke
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Borte
- Municipal Hospital "St. Georg" Children's Hospital, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany; Department of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Irina Lehmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany.
| | - Roland Eils
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Pharmacy and Molecular Biotechnology, and Bioquant Center, University of Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
| | - Saskia Trump
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany.
| |
Collapse
|
36
|
Koopmans T, Gosens R. Revisiting asthma therapeutics: focus on WNT signal transduction. Drug Discov Today 2017; 23:49-62. [PMID: 28890197 DOI: 10.1016/j.drudis.2017.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/20/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
Abstract
Asthma is a complex disease of the airways that develops as a consequence of both genetic and environmental factors. This interaction has highlighted genes important in early life, particularly those that control lung development, such as the Wingless/Integrase-1 (WNT) signalling pathway. Although aberrant WNT signalling is involved with an array of human conditions, it has received little attention within the context of asthma. Yet it is highly relevant, driving events involved with inflammation, airway remodelling, and airway hyper-responsiveness (AHR). In this review, we revisit asthma therapeutics by examining whether WNT signalling is a valid therapeutic target for asthma.
Collapse
Affiliation(s)
- Tim Koopmans
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, The Netherlands.
| |
Collapse
|
37
|
Prenatal noise stress impairs HPA axis and cognitive performance in mice. Sci Rep 2017; 7:10560. [PMID: 28874680 PMCID: PMC5585382 DOI: 10.1038/s41598-017-09799-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/28/2017] [Indexed: 11/11/2022] Open
Abstract
Noise stress is a common environmental pollutant whose adverse effect on offspring performance has been less studied. This study was novel in terms of using “noise” as a prenatal stress compared with physical stress to explore the effect of stress during gestation on HPA axis activation, cognitive performance, and motor coordination, as well as in investigating the effect of behavioral assessments on the corticosterone (CORT) levels. Three groups of C57BL/6 mice with a gestational history of either noise stress (NS), physical stress (PS), or no stress were examined in several behavioral tests. Plasma CORT level was significantly higher before starting the behavioral tests in NS group than the two other groups. It was significantly increased after the behavioral tests in both prenatal stressed groups relative to the controls. Stress caused anxiety-like behavior and reduced learning and memory performance in both stressed groups compared to the controls, as well as decreased motor coordination in the NS group relative to the other groups. The findings suggested that: prenatal NS severely changes the HPA axis; both prenatal stressors, and particularly NS, negatively impair the offspring’s cognitive and motor performance; and, they also cause a strong susceptibility to interpret environmental experiences as stressful conditions.
Collapse
|
38
|
Neuropsychiatry phenotype in asthma: Psychological stress-induced alterations of the neuroendocrine-immune system in allergic airway inflammation. Allergol Int 2017; 66S:S2-S8. [PMID: 28669635 DOI: 10.1016/j.alit.2017.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/11/2017] [Accepted: 05/14/2017] [Indexed: 01/11/2023] Open
Abstract
Since the recognition of asthma as a syndrome with complex pathophysiological signs and symptoms, recent research has sought to classify asthma phenotypes based on its clinical and molecular pathological features. Psychological stress was first recognized as a potential immune system modulator of asthma at the end of the 19th century. The activation of the central nervous system (CNS) upon exposure to psychological stress is integral for the initiation of signal transduction processes. The stress hormones, including glucocorticoids, epinephrine, and norepinephrine, which are secreted following CNS activation, are involved in the immunological alterations involved in psychological stress-induced asthma exacerbation. The mechanisms underlying this process may involve a pathological series of events from the brain to the lungs, which is attracting attention as a conceptually advanced phenotype in asthma pathogenesis. This review presents insights into the critical role of psychological stress in the development and exacerbation of allergic asthma, with a special focus on our own data that emphasizes on the continuity from the central sensing of psychological stress to enhanced eosinophilic airway inflammation.
Collapse
|
39
|
Association of prenatal and early childhood stress with reduced lung function in 7-year-olds. Ann Allergy Asthma Immunol 2017; 119:153-159. [PMID: 28668548 DOI: 10.1016/j.anai.2017.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/03/2017] [Accepted: 05/30/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND No prior study has examined associations between prenatal and early-life stress on childhood lung function or identified critical windows of exposure. OBJECTIVE To prospectively examine associations between prenatal and early-life stress and childhood lung function. METHODS Stress was indexed by a maternal negative life events (NLEs) score ascertained during pregnancy and between 1 and 2 years post partum. Spirometry was performed when children were a mean (SD) of 6.99 (0.89) years old. Associations of prenatal and early postnatal stress with spirometry z scores were examined in 199 children using linear regression. Effect modification by child sex was explored. RESULTS Most mothers were minorities (65% Hispanic, 21% African American), had 12 years or less of education (67%), and did not smoke prenatally (78%). The highest level of prenatal stress (≥5 NLEs) was associated with lower levels of forced expiratory volume in 1 second (FEV1) (z score = -0.53, P = .03), forced vital capacity (FVC) (z score = -0.49, P = .04), and forced expiratory flow between 25% and 75% (FEF25%-75%) (z score = -0.68, P = .01) after covariate adjustment; effects were similar for postnatal stress considered separately. In sex-stratified analyses, high postnatal stress (≥5 NLEs) was associated with lower FEV1 (z score = -0.76, P = .01), FVC (z score = -0.77, P = .01), and FEF25%-75% (z score = -0.67, P = .02) in boys but not girls, although the interaction term was not significant (P for interaction >.10). CONCLUSION These are the first prospective data that link perinatal stress with reduced child lung function. High levels of stress in the prenatal and postnatal periods were associated with symmetric reductions in FEV1 and FVC consistent with impaired lung growth. Given that lung function growth patterns are established by 7 years of age, these findings have lifelong implications.
Collapse
|
40
|
DeVries A, Vercelli D. The neonatal methylome as a gatekeeper in the trajectory to childhood asthma. Epigenomics 2017; 9:585-593. [PMID: 28322586 DOI: 10.2217/epi-2016-0149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Asthma is a heterogeneous group of conditions that typically begin in early life and result in recurrent, reversible bronchial obstruction. The role played by epigenetic mechanisms in the pathogenesis of childhood asthma is understood only in part. Here we discuss asthma epigenetics within a developmental perspective based on our recent demonstration that the epigenetic trajectory to childhood asthma begins at birth. We next discuss how this trajectory may be affected by prenatal environmental exposures. Finally, we examine in vitro studies that model the impact of asthma-associated exposures on the epigenome. All of these studies specifically surveyed human DNA methylation and involved a genome-wide component. In combination, their results broaden our understanding of asthma pathogenesis and the role the methylome plays in this process.
Collapse
Affiliation(s)
- Avery DeVries
- Graduate Program in Cellular & Molecular Medicine, University of Arizona, Tucson, AZ, USA.,Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Donata Vercelli
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.,Arizona Center for the Biology of Complex Diseases, University of Arizona, Tucson, AZ, USA.,Department of Cellular & Molecular Medicine, University of Arizona, Tucson, AZ, USA.,The Bio5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
41
|
Choi H, Song WM, Zhang B. Linking childhood allergic asthma phenotypes with endotype through integrated systems biology: current evidence and research needs. REVIEWS ON ENVIRONMENTAL HEALTH 2017; 32:55-63. [PMID: 28170342 DOI: 10.1515/reveh-2016-0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/11/2016] [Indexed: 06/06/2023]
Abstract
Asthma and other complex diseases results from a complex web of interactions involving inflammation, immunity, cell cycle, apoptosis, and metabolic perturbations across multiple organ systems. The extent to which various degrees of the age at onset, symptom severity, and the natural progression of the disease reflect multiple disease subtypes, influenced by unique process of development remains unknown. One of the most critical challenges to our understanding stems from incomplete understanding of the mechanisms. Within this review, we focus on the phenotypes of childhood allergic asthma as the basis to better understand the endotype for quantitative define subtypes of asthma. We highlight some of the known mechanistic pathways associated with the key hallmark events before the asthma onset. In particular, we examine how the recent advent of multiaxial -omics technologies and systems biology could help to clarify our current understanding of the pathway. We review how a large volume of molecular, genomic data generated by multiaxial technologies could be digested to identify cogent pathophysiologic molecular networks. We highlight some recent successes in application of these technologies within the context of other disease conditions for therapeutic interventions. We conclude by summarizing the research needs for the predictive value of preclinical biomarkers.
Collapse
|
42
|
Dewi DL, Mohapatra SR, Blanco Cabañes S, Adam I, Somarribas Patterson LF, Berdel B, Kahloon M, Thürmann L, Loth S, Heilmann K, Weichenhan D, Mücke O, Heiland I, Wimberger P, Kuhlmann JD, Kellner KH, Schott S, Plass C, Platten M, Gerhäuser C, Trump S, Opitz CA. Suppression of indoleamine-2,3-dioxygenase 1 expression by promoter hypermethylation in ER-positive breast cancer. Oncoimmunology 2017; 6:e1274477. [PMID: 28344890 DOI: 10.1080/2162402x.2016.1274477] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022] Open
Abstract
Kynurenine formation by tryptophan-catabolic indoleamine-2,3-dioxygenase 1 (IDO1) plays a key role in tumor immune evasion and inhibition of IDO1 is efficacious in preclinical models of breast cancer. As the response of breast cancer to immune checkpoint inhibitors may be limited, a better understanding of the expression of additional targetable immunomodulatory pathways is of importance. We therefore investigated the regulation of IDO1 expression in different breast cancer subtypes. We identified estrogen receptor α (ER) as a negative regulator of IDO1 expression. Serum kynurenine levels as well as tumoral IDO1 expression were lower in patients with ER-positive than ER-negative tumors and an inverse relationship between IDO1 and estrogen receptor mRNA was observed across 14 breast cancer data sets. Analysis of whole genome bisulfite sequencing, 450k, MassARRAY and pyrosequencing data revealed that the IDO1 promoter is hypermethylated in ER-positive compared with ER-negative breast cancer. Reduced induction of IDO1 was also observed in human ER-positive breast cancer cell lines. IDO1 induction was enhanced upon DNA demethylation in ER-positive but not in ER-negative cells and methylation of an IDO1 promoter construct reduced IDO1 expression, suggesting that enhanced methylation of the IDO1 promoter suppresses IDO1 in ER-positive breast cancer. The association of ER overexpression with epigenetic downregulation of IDO1 appears to be a particular feature of breast cancer as IDO1 was not suppressed by IDO1 promoter hypermethylation in the presence of high ER expression in cervical or endometrial cancer.
Collapse
Affiliation(s)
- Dyah L Dewi
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Soumya R Mohapatra
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Saioa Blanco Cabañes
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Isabell Adam
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | | | - Bianca Berdel
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Masroor Kahloon
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Loreen Thürmann
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ , Leipzig, Germany
| | - Stefanie Loth
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ , Leipzig, Germany
| | - Katharina Heilmann
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Dieter Weichenhan
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Oliver Mücke
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT Arctic University of Norway , Tromsø, Norway
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sarah Schott
- Department of Obstetrics and Gynecology, University of Heidelberg , Heidelberg, Germany
| | - Christoph Plass
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Michael Platten
- Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clarissa Gerhäuser
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Saskia Trump
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ , Leipzig, Germany
| | - Christiane A Opitz
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
43
|
Suh DI, Chang HY, Lee E, Yang SI, Hong SJ. Prenatal Maternal Distress and Allergic Diseases in Offspring: Review of Evidence and Possible Pathways. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2017; 9:200-211. [PMID: 28293926 PMCID: PMC5352571 DOI: 10.4168/aair.2017.9.3.200] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 12/14/2022]
Abstract
Recent studies have suggested a close association between prenatal maternal distress and allergic diseases in the offspring. We selected relevant birth-cohort or national registry studies using a keyword search of the PubMed database and summarized current evidence on the impact of prenatal maternal distress on the development of offspring's allergic diseases. Moreover, we postulated possible pathways linking prenatal distress and allergic diseases based on relevant human and animal studies. Both dysregulated hypothalamic-pituitary-adrenal axis and increased oxidative stress may cause structural (altered brain/lung development) and functional (skewed immune development) changes, which may predispose the fetus to developing allergic diseases during childhood. Although many facts are yet to be discovered, changes in the placental response and epigenetic modification are presumed to mediate the whole process from maternal distress to allergic diseases. Maternal prenatal distress can also interact with other physical or environmental factors, including familial or physical factors, indoor and outdoor pollutants, and early childhood psychological distress. The gut-microbiome-brain axis and the role of the microbiome as an immune modulator should be considered when investigating the stress-allergy relationship and exploring potential intervention modalities. Further research is needed, and particular attention should be given to defining the most vulnerable subjects and critical time periods. To this end, studies exploring relevant biomarkers are warranted, which can enable us to explore adequate intervention strategies.
Collapse
Affiliation(s)
- Dong In Suh
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Hyoung Yoon Chang
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Korea
| | - Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Gwangju, Korea
| | - Song I Yang
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Soo Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Environmental Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
44
|
Ota C, Baarsma HA, Wagner DE, Hilgendorff A, Königshoff M. Linking bronchopulmonary dysplasia to adult chronic lung diseases: role of WNT signaling. Mol Cell Pediatr 2016; 3:34. [PMID: 27718180 PMCID: PMC5055515 DOI: 10.1186/s40348-016-0062-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in infants caused by pre- and/or postnatal lung injury. BPD is characterized by arrested alveolarization and vascularization due to extracellular matrix remodeling, inflammation, and impaired growth factor signaling. WNT signaling is a critical pathway for normal lung development, and its altered signaling has been shown to be involved in the onset and progression of incurable chronic lung diseases in adulthood, such as chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF). In this review, we summarize the impact of WNT signaling on different stages of lung development and its potential contribution to developmental lung diseases, especially BPD, and chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Chiharu Ota
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.,The Perinatal Center, Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|