1
|
Fisher JJ, Grace T, Castles NA, Jones EA, Delforce SJ, Peters AE, Crombie GK, Hoedt EC, Warren KE, Kahl RG, Hirst JJ, Pringle KG, Pennell CE. Methodology for Biological Sample Collection, Processing, and Storage in the Newcastle 1000 Pregnancy Cohort: Protocol for a Longitudinal, Prospective Population-Based Study in Australia. JMIR Res Protoc 2024; 13:e63562. [PMID: 39546349 DOI: 10.2196/63562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Research in the developmental origins of health and disease provides compelling evidence that adverse events during the first 1000 days of life from conception can impact life course health. Despite many decades of research, we still lack a complete understanding of the mechanisms underlying some of these associations. The Newcastle 1000 Study (NEW1000) is a comprehensive, prospective population-based pregnancy cohort study based in Newcastle, New South Wales, Australia, that will recruit pregnant women and their partners at 11-14 weeks' gestation, with assessments at 20, 28, and 36 weeks; birth; 6 weeks; and 6 months, in order to provide detailed data about the first 1000 days of life to investigate the developmental origins of noncommunicable diseases. OBJECTIVE The study aims to provide a longitudinal multisystem approach to phenotyping, supported by robust clinical data and collection of biological samples in NEW1000. METHODS This manuscript describes in detail the large variety of samples collected in the study and the method of collection, storage, and utility of the samples in the biobank, with a particular focus on incorporation of the samples into emerging and novel large-scale "-omics" platforms, including the genome, microbiome, epigenome, transcriptome, fragmentome, metabolome, proteome, exposome, and cell-free DNA and RNA. Specifically, this manuscript details the methods used to collect, process, and store biological samples, including maternal, paternal, and fetal blood, microbiome (stool, skin, vaginal, oral), urine, saliva, hair, toenail, placenta, colostrum, and breastmilk. RESULTS Recruitment for the study began in March 2021. As of July 2024, 1040 women and 684 partners were enrolled, with 922 infants born. The NEW1000 biobank contains 24,357 plasma aliquots from ethylenediaminetetraacetic acid (EDTA) tubes, 5284 buffy coat aliquots, 4000 plasma aliquots from lithium heparin tubes, 15,884 blood serum aliquots, 2977 PAX RNA tubes, 26,595 urine sample aliquots, 2280 fecal swabs, 17,687 microbiome swabs, 2356 saliva sample aliquots, 1195 breastmilk sample aliquots, 4007 placental tissue aliquots, 2680 hair samples, and 2193 nail samples. CONCLUSIONS NEW1000 will generate a multigenerational, deeply phenotyped cohort with a comprehensive biobank of samples relevant to a large variety of analyses, including multiple -omics platforms. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/63562.
Collapse
Affiliation(s)
- Joshua J Fisher
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Tegan Grace
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Nathan A Castles
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Elizabeth A Jones
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Sarah J Delforce
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Alexandra E Peters
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Gabrielle K Crombie
- School of Life and Medical Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Emily C Hoedt
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Kirby E Warren
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Richard Gs Kahl
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Craig E Pennell
- School of Medicine and Public Health, College and Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| |
Collapse
|
2
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
3
|
Chen RJ, Nabila A, Gal Toth J, Stuhlmann H, Toth M. The chemokine XCL1 functions as a pregnancy hormone to program offspring innate anxiety. Brain Behav Immun 2024; 118:178-189. [PMID: 38428650 PMCID: PMC11044916 DOI: 10.1016/j.bbi.2024.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Elevated levels of cytokines in maternal circulation increase the offspring's risk for neuropsychiatric disease. Because of their low homeostatic levels, circulating maternal cytokines during normal pregnancies have not been considered to play a role in fetal brain development and offspring behavior. Here we report that the T/NK cell chemotactic cytokine XCL1, a local paracrine immune signal, can function as a pregnancy hormone and is required for the proper development of placenta and male offspring approach-avoidance behavior. We found that circulating XCL1 levels were at a low pregestational level throughout pregnancy except for a midgestational rise and fall. Blunted elevation in maternal plasma XCL1 in dams with a genetic 5HT1A receptor deficit or following neutralization by anti-XCL1 antibodies increased the expression of tissue damage associated factors in WT fetal placenta and led to increased innate anxiety and stress reactivity in the WT male offspring. Therefore, chemokines like XCL1 may act as pregnancy hormones to regulate placenta development and offspring emotional behavior.
Collapse
Affiliation(s)
- Rosa J Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anika Nabila
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Judit Gal Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi Stuhlmann
- Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miklos Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
4
|
Crombie GK, Palliser HK, Shaw JC, Hanley BA, Moloney RA, Hirst JJ. Prenatal Stress Induces Translational Disruption Associated with Myelination Deficits. Dev Neurosci 2023; 45:290-308. [PMID: 37004512 DOI: 10.1159/000530282] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Disruptions to neurodevelopment are known to be linked to behavioral disorders in childhood and into adulthood. The fetal brain is extremely vulnerable to stimuli that alter inhibitory GABAergic pathways and critical myelination processes, programing long-term neurobehavioral disruption. The maturation of the GABAergic system into the major inhibitory pathway in the brain and the development of oligodendrocytes into mature cells capable of producing myelin are integral components of optimal neurodevelopment. The current study aimed to elucidate prenatal stress-induced mechanisms that disrupt these processes and to delineate the role of placental pathways in these adverse outcomes. Pregnant guinea pig dams were exposed to prenatal stress with strobe light exposure for 2 h/day on gestational age (GA) 35, 40, 45, 50, 55, 60, and 65, and groups of fetuses and placentae were collected after the stress exposure on GA40, GA50, GA60, and GA69 (term). Fetal plasma, placental, and brain tissue were collected for allopregnanolone and cortisol quantification with ELISA. Relative mRNA expression of genes of specific pathways of interest was examined with real-time PCR in placental and hippocampal tissue, and myelin basic protein (MBP) was quantified immunohistochemically in the hippocampus and surrounding regions for assessment of mature myelin. Prenatal stress in mid-late gestation resulted in disruptions to the translational machinery responsible for the production of myelin and decreased myelin coverage in the hippocampus and surrounding regions. The male placenta showed an initial protective increase in allopregnanolone concentrations in response to maternal psychosocial stress. The male and female placentae had a sex-dependent increase in neurosteroidogenic enzymes at term following prenatal stress. Independent from exposure to prenatal stress, at gestational day 60 - a critical period for myelin development, the placentae of female fetuses had increased capability of preventing cortisol transfer to the fetus through expression of 11-beta-hydroxysteroid dehydrogenase types 1 and 2. The deficits early in the process of maturation of myelination indicate that the reduced myelination observed at childhood equivalence in previous studies begins in fetal life. This negative programing persists into childhood, potentially due to dysregulation of MBP translation processes. Expression patterns of neurosteroidogenic enzymes in the placenta at term following stress may identify at-risk fetuses that have been exposed to a stressful in utero environment.
Collapse
Affiliation(s)
- Gabrielle K Crombie
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Hannah K Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Julia C Shaw
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Bethany A Hanley
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Roisin A Moloney
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jonathan J Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
5
|
Dong Y, Weng J, Zhu Y, Sun D, He W, Chen Q, Cheng J, Zhu Y, Jiang Y. Transcriptomic profiling of the developing brain revealed cell-type and brain-region specificity in a mouse model of prenatal stress. BMC Genomics 2023; 24:86. [PMID: 36829105 PMCID: PMC9951484 DOI: 10.1186/s12864-023-09186-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Prenatal stress (PS) is considered as a risk factor for many mental disorders. PS-induced transcriptomic alterations may contribute to the functional dysregulation during brain development. Here, we used RNA-seq to explore changes of gene expression in the mouse fetal brain after prenatal exposure to chronic unpredictable mild stress (CUMS). RESULTS We compared the stressed brains to the controls and identified groups of significantly differentially expressed genes (DEGs). GO analysis on up-regulated DEGs revealed enrichment for the cell cycle pathways, while down-regulated DEGs were mostly enriched in the neuronal pathways related to synaptic transmission. We further performed cell-type enrichment analysis using published scRNA-seq data from the fetal mouse brain and revealed cell-type-specificity for up- and down-regulated DEGs, respectively. The up-regulated DEGs were highly enriched in the radial glia, while down-regulated DEGs were enriched in different types of neurons. Cell deconvolution analysis further showed altered cell fractions in the stressed brain, indicating accumulation of neuroblast and impaired neurogenesis. Moreover, we also observed distinct brain-region expression pattern when mapping DEGs onto the developing Allen brain atlas. The up-regulated DEGs were primarily enriched in the dorsal forebrain regions including the cortical plate and hippocampal formation. Surprisingly, down-regulated DEGs were found excluded from the cortical region, but highly expressed on various regions in the ventral forebrain, midbrain and hindbrain. CONCLUSION Taken together, we provided an unbiased data source for transcriptomic alterations of the whole fetal brain after chronic PS, and reported differential cell-type and brain-region vulnerability of the developing brain in response to environmental insults during the pregnancy.
Collapse
Affiliation(s)
- Yuhao Dong
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jie Weng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yueyan Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Daijing Sun
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Wei He
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Qi Chen
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jin Cheng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Ying Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
6
|
Kaki S, DeRosa H, Timmerman B, Brummelte S, Hunter RG, Kentner AC. Developmental Manipulation-Induced Changes in Cognitive Functioning. Curr Top Behav Neurosci 2023; 63:241-289. [PMID: 36029460 PMCID: PMC9971379 DOI: 10.1007/7854_2022_389] [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] [Indexed: 10/15/2022]
Abstract
Schizophrenia is a complex neurodevelopmental disorder with as-yet no identified cause. The use of animals has been critical to teasing apart the potential individual and intersecting roles of genetic and environmental risk factors in the development of schizophrenia. One way to recreate in animals the cognitive impairments seen in people with schizophrenia is to disrupt the prenatal or neonatal environment of laboratory rodent offspring. This approach can result in congruent perturbations in brain physiology, learning, memory, attention, and sensorimotor domains. Experimental designs utilizing such animal models have led to a greatly improved understanding of the biological mechanisms that could underlie the etiology and symptomology of schizophrenia, although there is still more to be discovered. The implementation of the Research and Domain Criterion (RDoC) has been critical in taking a more comprehensive approach to determining neural mechanisms underlying abnormal behavior in people with schizophrenia through its transdiagnostic approach toward targeting mechanisms rather than focusing on symptoms. Here, we describe several neurodevelopmental animal models of schizophrenia using an RDoC perspective approach. The implementation of animal models, combined with an RDoC framework, will bolster schizophrenia research leading to more targeted and likely effective therapeutic interventions resulting in better patient outcomes.
Collapse
Affiliation(s)
- Sahith Kaki
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Holly DeRosa
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
- University of Massachusetts Boston, Boston, MA, USA
| | - Brian Timmerman
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University, Detroit, MI, USA
| | | | - Amanda C Kentner
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
7
|
Barroca NCB, Della Santa G, Suchecki D, García-Cairasco N, Umeoka EHDL. Challenges in the use of animal models and perspectives for a translational view of stress and psychopathologies. Neurosci Biobehav Rev 2022; 140:104771. [PMID: 35817171 DOI: 10.1016/j.neubiorev.2022.104771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 12/25/2022]
Abstract
The neurobiology and development of treatments for stress-related neuropsychiatric disorders rely heavily on animal models. However, the complexity of these disorders makes it difficult to model them entirely, so only specific features of human psychopathology are emulated and these models should be used with great caution. Importantly, the effects of stress depend on multiple factors, like duration, context of exposure, and individual variability. Here we present a review on pre-clinical studies of stress-related disorders, especially those developed to model posttraumatic stress disorder, major depression, and anxiety. Animal models provide relevant evidence of the underpinnings of these disorders, as long as face, construct, and predictive validities are fulfilled. The translational challenges faced by scholars include reductionism and anthropomorphic/anthropocentric interpretation of the results instead of a more naturalistic and evolutionary understanding of animal behavior that must be overcome to offer a meaningful model. Other limitations are low statistical power of analysis, poor evaluation of individual variability, sex differences, and possible conflicting effects of stressors depending on specific windows in the lifespan.
Collapse
Affiliation(s)
- Nayara Cobra Barreiro Barroca
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Giovanna Della Santa
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Norberto García-Cairasco
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil; Department of Physiology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Eduardo Henrique de Lima Umeoka
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil; School of Medicine, University Center UniCerrado, Goiatuba, GO, Brazil
| |
Collapse
|
8
|
Lipner E, Murphy SK, Breen EC, Cohn BA, Krigbaum NY, Cirillo PM, Alloy LB, Ellman LM. Infection and higher cortisol during pregnancy and risk for depressive symptoms in adolescent offspring. Psychoneuroendocrinology 2022; 141:105755. [PMID: 35429699 PMCID: PMC9149123 DOI: 10.1016/j.psyneuen.2022.105755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 02/24/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Prenatal infection, particularly at mid-gestation, has been associated with various psychopathological outcomes in offspring; however, findings linking prenatal infection to offspring depression outcomes have been mixed. Previous research indicates that it may be the co-occurrence of prenatal adversities (e.g., infection and stress) that are associated with depression outcomes in offspring. Nevertheless, no study to date has investigated whether higher levels of biomarkers linked to prenatal stress (e.g., cortisol) in the presence of infection may account for these outcomes. Participants were drawn from the Child Health and Development Studies (CHDS), a prospective, longitudinal study of pregnant women and their offspring. The present study included mother-offspring dyads from the Adolescent Study, a subsample of the CHDS cohort, whose offspring were assessed in adolescence and whose mothers also provided sera to be assayed for cortisol (n = 695). Hierarchical multivariable regressions were conducted to examine whether maternal cortisol during the first and second trimesters of pregnancy interacted with maternal infection to predict increased risk for symptoms of depression in adolescent offspring. There was a significant interaction of second trimester infection and higher cortisol on offspring depression scores during adolescence, controlling for maternal education (p = 0.04). Findings suggest that higher maternal cortisol may sensitize mothers and their offspring to the disruptive influences of infection during mid-pregnancy, conferring greater risk of depressive symptomatology in offspring.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Shannon K Murphy
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Elizabeth C Breen
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, University of California, 300 Medical Plaza, Los Angeles, CA 90095, USA
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Lauren B Alloy
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Lauren M Ellman
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA.
| |
Collapse
|
9
|
Haq SU, Bhat UA, Kumar A. Prenatal stress effects on offspring brain and behavior: Mediators, alterations and dysregulated epigenetic mechanisms. J Biosci 2021. [DOI: 10.1007/s12038-021-00153-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
10
|
Mareckova K, Miles A, Andryskova L, Brazdil M, Nikolova YS. Temporally and sex-specific effects of maternal perinatal stress on offspring cortical gyrification and mood in young adulthood. Hum Brain Mapp 2020; 41:4866-4875. [PMID: 33010202 PMCID: PMC7643354 DOI: 10.1002/hbm.25163] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/28/2020] [Indexed: 01/19/2023] Open
Abstract
Maternal stress during pregnancy and shortly thereafter is associated with altered offspring brain development that may increase risk of mood and anxiety disorders. Cortical gyrification is established during the prenatal period and the first 2 years of life and is altered in psychiatric disorders. Here, we sought to characterize the effects of perinatal stress exposure on offspring gyrification patterns and mood dysregulation in young adulthood. Participants included 85 young adults (56.5% women; 23–24 years) from the European Longitudinal Study of Pregnancy and Childhood (ELSPAC) with perinatal stress data across four distinct timepoints and structural MRI data from young adulthood. Perinatal stress exposure was measured as maternal stress during first and second half of pregnancy, first 6 months, and 6–18 months after birth. Cortical gyrification and mood dysregulation were quantified using local gyrification index (LGI), computed with Freesurfer, and the Profile of Mood States questionnaire, respectively. Perinatal stress predicted cortical gyrification in young adulthood, and its timing influenced location, direction, and sex‐specificity of effects. In particular, whereas early prenatal stress was associated with sex‐dependent medium‐to‐large effects in large temporal, parietal, and occipital regions (f2 = 0.19–0.38, p < .001), later perinatal stress was associated with sex‐independent small‐to‐medium effects in smaller, more anterior regions (f2 = 0.10–0.19, p < .003). Moreover, in females, early prenatal stress predicted higher LGI in a large temporal region, which was further associated with mood disturbance in adulthood (r = 0.399, p = .006). These findings point out the long‐term implications of perinatal stress exposure for cortical morphology and mood dysregulation.
Collapse
Affiliation(s)
- Klara Mareckova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Amy Miles
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Lenka Andryskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Milan Brazdil
- Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Blanc J, Eugene D, Louis EF, Cadichon JM, Joseph J, Pierre A, Laine R, Alexandre M, Huang KY. Mental Health Among Children Older than 10 Years Exposed to the Haiti 2010 Earthquake: a Critical Review. Curr Psychiatry Rep 2020; 22:57. [PMID: 32876808 PMCID: PMC8020445 DOI: 10.1007/s11920-020-01178-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW There is evidence that the exposure to a major natural disaster such as the 2010 earthquake in Haiti may have devastating short- and long-term effects on children's mental health and overall development. This review summarizes what has been reported in the past 3 years (2017-2020) regarding emotional, psychological, and behavioral effects of exposure to this particular earthquake on children 10 years and older. RECENT FINDINGS Twenty-six articles were screened, of which twenty-one were selected for final analysis. The main themes addressed in the literature over the 3 years concerned prevalence of post-traumatic stress disorder (PTSD), depression, determinants of resilience factors, prevalence of sexual violence as well as prenatal exposure, and subsequent autism behaviors/symptoms. The majority of the findings analyzed in this review on mental health in young Haitian survivors of the 2010 earthquake came from cross-sectional studies conducted in West region of Haiti, spefically Port-au-Prince. There was a paucity of longitudinal and translational data available. The results of this critical review can be used to inform disaster preparedness programs with the aim of protecting children's development and mental health, which are much needed on this disaster-prone island.
Collapse
Affiliation(s)
- Judite Blanc
- Division of Health and Behavior, Department of Population Health, Center for Healthful Behavior Change, New York University Grossman School of Medicine, 180 Madison Avenue, New York, NY, 10016, USA.
| | - Dominique Eugene
- Fogarty International Center and National Institute of Mental Health of the National Institutes of Health, Harvard University, Cambridge, USA
- Children's System of Care, Monterey County Behavioral Health Bureau, Salinas, CA, USA
| | - Elizabeth Farrah Louis
- Fogarty International Center and National Institute of Mental Health of the National Institutes of Health, Harvard University, Cambridge, USA
| | | | | | | | - Roudly Laine
- Université d'État d'Haïti, Port-au-Prince, Haiti
| | - Margarett Alexandre
- Department of Nursing, York College City University of New York, New York, USA
| | - Keng-Yen Huang
- Population Health and Child and Adolescent Psychiatry Center for Early Childhood Health & Development (CEHD), Department of Population Health, New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
12
|
Bush NR, Wakschlag LS, LeWinn KZ, Hertz-Picciotto I, Nozadi SS, Pieper S, Lewis J, Biezonski D, Blair C, Deardorff J, Neiderhiser JM, Leve LD, Elliott AJ, Duarte CS, Lugo-Candelas C, O’Shea TM, Avalos LA, Page GP, Posner J. Family Environment, Neurodevelopmental Risk, and the Environmental Influences on Child Health Outcomes (ECHO) Initiative: Looking Back and Moving Forward. Front Psychiatry 2020; 11:547. [PMID: 32636769 PMCID: PMC7318113 DOI: 10.3389/fpsyt.2020.00547] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
The family environment, with all its complexity and diverse components, plays a critical role in shaping neurodevelopmental outcomes in children. Herein we review several domains of the family environment (family socioeconomic status, family composition and home environment, parenting behaviors and interaction styles, parental mental health and functioning, and parental substance use) and discuss how these domains influence neurodevelopment, with particular emphasis on mental health outcomes. We also highlight a new initiative launched by the National Institutes of Health, the Environmental influences on Child Health Outcomes (ECHO) program. We discuss the role that ECHO will play in advancing our understanding of the impact of the family environment on children's risk for psychiatric outcomes. Lastly, we conclude with important unanswered questions and controversies in this area of research, highlighting how ECHO will contribute to resolving these gaps in our understanding, clarifying relationships between the family environment and children's mental health.
Collapse
Affiliation(s)
- Nicole R. Bush
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, CA, United States
| | - Lauren S. Wakschlag
- Department of Medical Social Sciences and Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, Il, United States
| | - Kaja Z. LeWinn
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA, United States
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis, Davis, CA, United States
| | - Sara S. Nozadi
- Community Environmental Health Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Sarah Pieper
- Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, United States
| | - Johnnye Lewis
- Community Environmental Health Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Dominik Biezonski
- Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, United States
| | - Clancy Blair
- Department of Population Health, New York University, New York, NY, United States
| | - Julianna Deardorff
- Community Health Sciences, University of California, Berkeley, Berkeley, CA, United States
| | - Jenae M. Neiderhiser
- Department of Psychology, Penn State University, University Park, PA, United States
| | - Leslie D. Leve
- Prevention Science Institute, University of Oregon, Eugene, OR, United States
| | - Amy J. Elliott
- Center for Pediatric and Community Research, Avera Research Institute, Sioux Falls, SD, United States
| | - Cristiane S. Duarte
- Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, United States
| | - Claudia Lugo-Candelas
- Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, United States
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lyndsay A. Avalos
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, United States
| | - Grier P. Page
- Department of Biostatistics and Epidemiology, RTI, Atlanta, GA, United States
| | - Jonathan Posner
- Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, United States
| |
Collapse
|
13
|
Marecková K, Klasnja A, Bencurova P, Andrýsková L, Brázdil M, Paus T. Prenatal Stress, Mood, and Gray Matter Volume in Young Adulthood. Cereb Cortex 2020; 29:1244-1250. [PMID: 29425268 DOI: 10.1093/cercor/bhy030] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 01/19/2018] [Indexed: 01/22/2023] Open
Abstract
This study aimed to determine whether prenatal stress, measured by the number of stressful life events during the first 20 weeks of pregnancy, might relate to mood dysregulation and altered brain structure in young adulthood. Participants included 93 young adults from a community-based birth cohort from the Czech Republic. Information on prenatal stress exposure was collected from their mothers in 1990-1992. Magnetic resonance imaging (MRI) and mood-related data were collected from the young adults in 2015. MRI analyses focused on overall gray matter (GM) volume and GM volume of cortical regions previously associated with major depression. Higher prenatal stress predicted more mood dysregulation, lower overall GM volume, and lower GM volume in mid-dorsolateral frontal cortex, anterior cingulate cortex, and precuneus in young adulthood. We observed no prenatal stress by sex interactions for any of the relations. We conclude that prenatal stress is an important risk factor that relates to worse mood states and altered brain structure in young adulthood irrespective of sex. Our results point to the importance and long-lasting effects of prenatal programming and suggest that offspring of mothers who went through substantial stress during pregnancy might benefit from early intervention that would reduce the odds of mental illness in later life.
Collapse
Affiliation(s)
- Klára Marecková
- Brain and Mind Research Programme, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Anja Klasnja
- Rotman Research Institute, Baycrest, Toronto, ON, Canada
| | - Petra Bencurova
- Brain and Mind Research Programme, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Andrýsková
- Research Centre for Toxic Compounds in the Environment (RECETOX), Faculty of Science, MU, Brno, Czech Republic
| | - Milan Brázdil
- Brain and Mind Research Programme, Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Neurology, St. Anne's University Hospital and Faculty of medicine, MU, Brno, Czech Republic
| | - Tomáš Paus
- Rotman Research Institute, Baycrest, Toronto, ON, Canada.,Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada.,Child Mind Institute, New York, NY, USA
| |
Collapse
|
14
|
Ellman LM, Murphy SK, Maxwell SD, Calvo EM, Cooper T, Schaefer CA, Bresnahan MA, Susser ES, Brown AS. Maternal cortisol during pregnancy and offspring schizophrenia: Influence of fetal sex and timing of exposure. Schizophr Res 2019; 213:15-22. [PMID: 31345704 PMCID: PMC7074891 DOI: 10.1016/j.schres.2019.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Maternal stress during pregnancy has been repeatedly linked to increased risk for schizophrenia; however, no study has examined maternal cortisol during pregnancy and risk for the disorder. Study aims were to determine whether prenatal cortisol was associated with risk for schizophrenia and risk for an intermediate phenotype-decreased fetal growth-previously linked to prenatal cortisol and schizophrenia. Timing of exposure and fetal sex also were examined given previous findings. METHODS Participants were 64 cases diagnosed with schizophrenia spectrum disorders (SSD) and 117 controls from a prospective birth cohort study. Maternal cortisol was determined from stored sera from each trimester and psychiatric diagnoses were assessed from offspring using semi-structured interviews and medical records review. RESULTS Maternal cortisol during pregnancy was not associated with risk for offspring schizophrenia. There was a significant interaction between 3rd trimester cortisol and case status on fetal growth. Specifically, cases exposed to higher 3rd trimester maternal cortisol had significantly decreased fetal growth compared to controls. In addition, these findings were restricted to male offspring. CONCLUSIONS Our results indicate that higher prenatal cortisol is associated with an intermediate phenotype linked to schizophrenia, fetal growth, but only among male offspring who developed schizophrenia. Findings were consistent with evidence that schizophrenia genes may disrupt placental functioning specifically for male fetuses, as well as findings that males are more vulnerable to maternal cortisol during pregnancy. Finally, results suggest that examining fetal sex and intermediate phenotypes may be important in understanding the mechanisms involved in prenatal contributors to schizophrenia.
Collapse
Affiliation(s)
- Lauren M Ellman
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Shannon K Murphy
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Seth D Maxwell
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Evan M Calvo
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Thomas Cooper
- Analytic Psychopharmacology, Nathan S. Kline Institute, 140 Old Orangeburg Road Orangeburg, NY 10962, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America; Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Catherine A Schaefer
- Division of Research, Kaiser Permanente, 2000 Broadway, Oakland, CA 94612, United States of America.
| | - Michaeline A Bresnahan
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Ezra S Susser
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Alan S Brown
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America; Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States of America.
| |
Collapse
|
15
|
Atrooz F, Liu H, Salim S. Stress, psychiatric disorders, molecular targets, and more. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:77-105. [PMID: 31601407 DOI: 10.1016/bs.pmbts.2019.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mental health is central to normal health outcomes. A widely accepted theory is that chronic persistent stress during adulthood as well as during early life triggers onset of neuropsychiatric ailments. However, questions related to how that occurs, and why are some individuals resistant to stress while others are not, remain unanswered. An integrated, multisystemic stress response involving neuroinflammatory, neuroendocrine, epigenetic and metabolic cascades have been suggested to have causative links. Several theories have been proposed over the years to conceptualize this link including the cytokine hypothesis, the endocrine hypothesis, the oxidative stress hypothesis and the oxido-neuroinflammation hypothesis. The data discussed in this review describes potential biochemical basis of the link between stress, and stress-induced neuronal, behavioral and emotional deficits, providing insights into potentially novel drug targets.
Collapse
Affiliation(s)
- Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Hesong Liu
- Baylor College of Medicine, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.
| |
Collapse
|
16
|
Abstract
Women undergo developmental and cyclic changes in hormonal exposures that affect brain function and mental health. Some women are more vulnerable to the effects of these hormonal exposures, for reasons that remain to be determined. Evidence to date indicates that anxiety and mood disorders are the most sensitive to hormonal fluctuations in women but there is also growing evidence for a protective effect of female reproductive hormones on schizophrenia. The hormonal exposures of the menstrual cycle, pregnancy, the postpartum period, lactation, and menopause are quite different and may be associated with at least partially distinct symptom profiles.
Collapse
Affiliation(s)
- Margaret Altemus
- VA Connecticut Health Care System, Women's Clinic, Building 2, Room 7-165, 950 Campbell Avenue, New Haven, CT 06516, USA.
| |
Collapse
|
17
|
Jones SL, Dufoix R, Laplante DP, Elgbeili G, Patel R, Chakravarty MM, King S, Pruessner JC. Larger Amygdala Volume Mediates the Association Between Prenatal Maternal Stress and Higher Levels of Externalizing Behaviors: Sex Specific Effects in Project Ice Storm. Front Hum Neurosci 2019; 13:144. [PMID: 31156408 PMCID: PMC6528106 DOI: 10.3389/fnhum.2019.00144] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/15/2019] [Indexed: 01/18/2023] Open
Abstract
Introduction: The amygdala is a brain structure involved in emotional regulation. Studies have shown that larger amygdala volumes are associated with behavioral disorders. Prenatal maternal depression is associated with structural changes in the amygdala, which in turn, is predictive of an increase in behavioral problems. Girls may be particularly vulnerable. However, it is not known whether disaster-related prenatal maternal stress (PNMS), or which aspect of the maternal stress experience (i.e., objective hardship, subjective distress, and cognitive appraisal), influences amygdala volumes. Nor is it known whether amygdala volumes mediate the effect of PNMS on behavioral problems in girls and boys. Aims: To assess whether aspects of PNMS are associated with amygdala volume, to determine whether timing of exposure moderates the effect, and to test whether amygdala volume mediates the association between PNMS and internalizing and externalizing problems in 11½ year old children exposed in utero, to varying levels of disaster-related PNMS. Methods: Bilateral amygdala volumes (AGV) and total brain volume (TBV) were acquired using magnetic resonance imaging, from 35 boys and 33 girls whose mothers were pregnant during the January 1998 Quebec Ice Storm. The mothers' disaster-related stress was assessed in June 1998. Child internalizing and externalizing problems were assessed at 11½ years using the Child Behavior Checklist (CBCL). Hierarchical regression analyses and mediation analyses were conducted on boys and girls separately, controlling for perinatal and postnatal factors. Results: In boys, subjective distress was associated with larger right AGV/TBV when mothers where exposed during late pregnancy, which in turn explained higher levels of externalizing behavior. However, when adjusting for postnatal factors, the effect was no longer significant. In girls, later gestational exposure to the ice storm was associated with larger AGV/TBV, but here, higher levels of objective PNMS were associated with more externalizing problems, which was, in part, mediated by larger AGV/TBV. No effects were detected on internalizing behaviors. Conclusion: These results suggest that the effects of PNMS on amygdala development and externalizing symptoms, as assessed in boys and girls in early adolescence, can be influenced by the timing of the stress in pregnancy, and the particular aspect of the mother's stress experience.
Collapse
Affiliation(s)
- Sherri Lee Jones
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Romane Dufoix
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - David P. Laplante
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Guillaume Elgbeili
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Raihaan Patel
- Computational Brain Anatomy Laboratory (CoBrA Lab), Douglas Mental Health University Institute, Montreal, QC, Canada
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - M. Mallar Chakravarty
- Computational Brain Anatomy Laboratory (CoBrA Lab), Douglas Mental Health University Institute, Montreal, QC, Canada
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Suzanne King
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Jens C. Pruessner
- Laboratory of Jens Pruessner, Department of Psychology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
18
|
Kolaka R, Chotwiwatthanakun C, Chutabhakdikul N. Fetal exposure to high levels of maternal glucocorticoids alters reelin signaling in the prefrontal cortex of rat pups. Int J Dev Neurosci 2019; 78:185-190. [PMID: 31014819 DOI: 10.1016/j.ijdevneu.2019.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022] Open
Abstract
Maternal stress (MS) is associated with various neuropsychiatric disorders and cognitive impairment in the offspring. However, it is unclear how early life stress alters the pup's brain development and how it contributes to the pathology of neuropsychiatric disorders later in life. Reelin is a large extracellular matrix glycoprotein that plays essential roles in early brain development such as neural migration, synaptic development, and maturation. Dysregulation of reelin and its signaling proteins is associated with the emergence of neuropsychiatric disorders in adulthood. This study examined the effect of repeated maternal Carbenoxolone (CBX) injection during late gestation on reelin signaling in the prefrontal cortex (PFC) of rat pups. CBX is a selective 11β-HSD2 enzyme inhibitor that promotes the direct transfer of maternal corticosteroids (CORT) to the fetus. Therefore, treatment with CBX can mimic the animal model of early life exposure to high levels of maternal stress hormone. In this study, pregnant rats were injected daily with either saline or CBX during gestation day (GD) 14-21, and the levels of reelin and its signaling proteins were examined in the PFC of rat pups at different postnatal age from P0-P21. The main result of this study is the repeated maternal CBX injections during GD14-21 acutely increase reln mRNA and protein expression in the PFC of rat pups at birth (P0) and follow by a significant decrease during P7-P14. The treatment also causes long term decreases in the amount of VLDLR and Dab1 which are the downstream signaling proteins for the reelin pathway, at least until P21. Our results indicated that fetal exposure to high levels of maternal CORT interferes with reelin signaling which might have profound effects on cortical development associated with neuropsychiatric disorders later in life.
Collapse
Affiliation(s)
- Ratirat Kolaka
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | | | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| |
Collapse
|
19
|
Conway F, Brown AS. Maternal Immune Activation and Related Factors in the Risk of Offspring Psychiatric Disorders. Front Psychiatry 2019; 10:430. [PMID: 31316403 PMCID: PMC6611212 DOI: 10.3389/fpsyt.2019.00430] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/31/2019] [Indexed: 12/25/2022] Open
Abstract
Maternal immune activation (MIA) at the time of gestation has been linked to increased risk of neurodevelopmental psychiatric disorders. Animal and human models have been used to evaluate the relationship between MIA and these outcomes. Given that each of these two disciplines of study have their benefits and limitations, a translational perspective is expected to illuminate more than by the use of any single approach. In this article, we discuss this translational framework and explore how it may be enhanced by the utilization of epigenetic studies and by investigating the microbiome. In this perspectives piece, we focus on the impact of epidemiologic studies, animal models, and preclinical studies in the literature on MIA as well as the potential for greater integration between fields.
Collapse
Affiliation(s)
- Fiona Conway
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| | - Alan S Brown
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
20
|
Haselbeck C, Niederberger U, Gubi-Kelm S, Jahn F, Dautwiz F, Siniatchkin M. Secure attachment style appears to compensate for the effect of prenatal maternal distress regarding difficult infant temperament development. ZEITSCHRIFT FUR KINDER-UND JUGENDPSYCHIATRIE UND PSYCHOTHERAPIE 2018; 47:239-251. [PMID: 30080118 DOI: 10.1024/1422-4917/a000606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective: Secure attachment style is a known protective factor regarding psychopathological development. The infant's attachment style, which is developed during the first two years of life, is therefore considered a moderating factor on the association between prenatal maternal distress and child temperament development which has repeatedly been reported in previous studies. Method: In this longitudinal study on a new sample of 51 mother-child-dyads, reported maternal distress and maternal empathy were assessed during pregnancy. Infant temperament and motor development were assessed at 12 months, while additionally controlling for the infant's attachment style as a postnatal factor. Results: Infants with secure attachment style whose mothers had experienced higher prenatal distress showed slightly better gross motor development at the age of 12 months. No association could be found between prenatal maternal distress and infant temperament. Conclusions: The results support the view that secure attachment style in children is a protective factor and softens the effects of prenatal maternal distress on difficult temperament development.
Collapse
Affiliation(s)
- Christin Haselbeck
- 1 Institut für Medizinische Psychologie und Medizinische Soziologie, Universitätsklinikum Schleswig-Holstein, Campus, Kiel, Germany
| | - Uwe Niederberger
- 1 Institut für Medizinische Psychologie und Medizinische Soziologie, Universitätsklinikum Schleswig-Holstein, Campus, Kiel, Germany
| | - Silvia Gubi-Kelm
- 2 Department of Psychology, Medical School Hamburg, Hamburg, Germany
| | - Franziska Jahn
- 1 Institut für Medizinische Psychologie und Medizinische Soziologie, Universitätsklinikum Schleswig-Holstein, Campus, Kiel, Germany
| | - Friederike Dautwiz
- 1 Institut für Medizinische Psychologie und Medizinische Soziologie, Universitätsklinikum Schleswig-Holstein, Campus, Kiel, Germany
| | - Michael Siniatchkin
- 1 Institut für Medizinische Psychologie und Medizinische Soziologie, Universitätsklinikum Schleswig-Holstein, Campus, Kiel, Germany
| |
Collapse
|
21
|
Perinatal stress and human hippocampal volume: Findings from typically developing young adults. Sci Rep 2018; 8:4696. [PMID: 29549289 PMCID: PMC5856850 DOI: 10.1038/s41598-018-23046-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/05/2018] [Indexed: 12/27/2022] Open
Abstract
The main objective of this study was to investigate the impact of prenatal and early postnatal stress on hippocampal volume in young adulthood. In sharp contrast to numerous results in animal models, our data from a neuroimaging follow-up (n = 131) of a community-based birth cohort from the Czech Republic (European Longitudinal Study of Pregnancy and Childhood) showed that in typically developing young adults, hippocampal volume was not associated with birth weight, stressful life events during the prenatal or early postnatal period, or dysregulated mood and wellbeing in the mother during the early postnatal period. Interestingly, mother’s anxiety/co-dependence during the first weeks after birth did show long-lasting effects on the hippocampal volume in young adult offspring irrespective of sex. Further analyses revealed that these effects were subfield-specific; present in CA1, CA2/3, CA4, GC-DG, subiculum, molecular layer, and HATA, hippocampal subfields identified by translational research as most stress- and glucocorticoid-sensitive, but not in the remaining subfields. Our findings provide evidence that the type of early stress is critical when studying its effects on the human brain.
Collapse
|
22
|
Maxwell SD, Fineberg AM, Drabick DA, Murphy SK, Ellman LM. Maternal Prenatal Stress and Other Developmental Risk Factors for Adolescent Depression: Spotlight on Sex Differences. JOURNAL OF ABNORMAL CHILD PSYCHOLOGY 2018; 46:381-397. [PMID: 28393324 PMCID: PMC5828524 DOI: 10.1007/s10802-017-0299-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maternal stress during pregnancy has been linked to premorbid abnormalities associated with depression (e.g., difficult temperament, cognitive deficits) in offspring. However, few studies have looked across developmental periods to examine maternal stress during pregnancy and offspring depression during adolescence and whether these associations differ by sex. The current study used data from 1711 mother-offspring dyads (offspring sex: 49.8% male) in a longitudinal birth cohort study. Maternal narratives collected during pregnancy were qualitatively coded for stress-related themes by independent raters. Latent class analysis (LCA) identified distinct subgroups of offspring based on exposure to maternal prenatal stress and other developmental factors from the prenatal, childhood, and adolescent periods that have been associated with depression and/or maternal prenatal stress. LCA identified subgroups that were compared to determine whether and to what extent they differed on adolescent depressive symptoms. LCA revealed a subgroup of "high-risk" individuals, characterized by maternal factors during pregnancy (higher ambivalence/negativity and lower positivity towards the pregnancy, higher levels of hassles, lower maternal education and higher maternal age at birth, higher pre-pregnancy BMI) and offspring developmental factors (decreased cognitive functioning during childhood and adolescence, lower perceived parental support during adolescence, and higher levels of maternal depression during adolescence). High-risk females exhibited elevated conduct symptoms and higher birth order, while high-risk males exhibited decreased internalizing symptoms and lower birth order. Both high-risk males and females reported elevated depressive symptoms during adolescence relative to their "low-risk" counterparts.
Collapse
Affiliation(s)
- Seth D Maxwell
- Department of Psychology, Temple University, 1701 N. 13th Street, Philadelphia, PA, 19122, USA
| | - Anna M Fineberg
- Department of Psychology, Temple University, 1701 N. 13th Street, Philadelphia, PA, 19122, USA
| | - Deborah A Drabick
- Department of Psychology, Temple University, 1701 N. 13th Street, Philadelphia, PA, 19122, USA
| | - Shannon K Murphy
- Department of Psychology, Temple University, 1701 N. 13th Street, Philadelphia, PA, 19122, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, 1701 N. 13th Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
23
|
Boschen KE, Keller SM, Roth TL, Klintsova AY. Epigenetic mechanisms in alcohol- and adversity-induced developmental origins of neurobehavioral functioning. Neurotoxicol Teratol 2018; 66:63-79. [PMID: 29305195 DOI: 10.1016/j.ntt.2017.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
The long-term effects of developmental alcohol and stress exposure are well documented in both humans and non-human animal models. Damage to the brain and attendant life-long impairments in cognition and increased risk for psychiatric disorders are debilitating consequences of developmental exposure to alcohol and/or psychological stress. Here we discuss evidence for a role of epigenetic mechanisms in mediating these consequences. While we highlight some of the common ways in which stress or alcohol impact the epigenome, we point out that little is understood of the epigenome's response to experiencing both stress and alcohol exposure, though stress is a contributing factor as to why women drink during pregnancy. Advancing our understanding of this relationship is of critical concern not just for the health and well-being of individuals directly exposed to these teratogens, but for generations to come.
Collapse
Affiliation(s)
- K E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, United States
| | - S M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - T L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - A Y Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
24
|
Abstract
Over the last few decades, evidence has emerged that the pathogenesis of psychiatric disorders such as schizophrenia can involve perturbations of the hypothalamic-pituitary-adrenal (HPA) axis and other neuroendocrine systems. Variations in the manifestation of these effects could be related to differences in clinical symptoms between affected individuals and to differences in treatment response. Such effects can also arise from the complex interaction between genes and environmental factors. Here, we review the effects of maternal stress on abnormalities in HPA axis regulation and the development of psychiatric disorders such as schizophrenia. Studies in this area may prove critical for increasing our understanding of the multidimensional nature of mental disorders and could lead to the development of improved diagnostics and novel therapeutic approaches for treating individuals who suffer from these conditions.
Collapse
Affiliation(s)
| | - Paul C Guest
- Laboratory of Neuroproteomics, Institute of Biology, University of Campinas, Campinas, Brazil.
| |
Collapse
|
25
|
Murphy SK, Fineberg AM, Maxwell SD, Alloy LB, Zimmermann L, Krigbaum NY, Cohn BA, Drabick DAG, Ellman LM. Maternal infection and stress during pregnancy and depressive symptoms in adolescent offspring. Psychiatry Res 2017; 257:102-110. [PMID: 28750213 PMCID: PMC5823248 DOI: 10.1016/j.psychres.2017.07.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/26/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
Maternal infection during pregnancy has been linked to increased risk of offspring depression. Additionally, maternal stress during pregnancy has been consistently linked with adverse offspring outcomes associated with depression. Relatedly, stress has been associated with increased risk of infection; however no study has investigated stress-infection interactions during pregnancy and risk for offspring depression. Participants were drawn from the Child Health and Development Studies (CHDS), a prospective, longitudinal study that enrolled pregnant women from 1959 to 1966. Maternal health and birth outcome information were collected, as well as open-ended interviews about worrisome events during pregnancy. The present study included participants from a subsample of women whose offspring (n = 1711) completed self-reports of depressive symptoms during adolescence. Results indicated that maternal infection during only the second trimester was associated with higher scores on adolescent offspring depressive symptoms, while controlling for maternal education at birth, adolescent age, and maternal depressive symptoms at adolescence. Maternal experiences of daily stress during pregnancy moderated this association, such that mothers diagnosed with second trimester infection and who experienced daily stress had offspring with significantly higher depression scores than mothers of adolescents diagnosed with an infection alone. Findings have potential implications for prevention and intervention strategies.
Collapse
Affiliation(s)
| | - Anna M. Fineberg
- Temple University, Department of Psychology, Philadelphia, PA, USA
| | - Seth D. Maxwell
- Temple University, Department of Psychology, Philadelphia, PA, USA
| | - Lauren B. Alloy
- Temple University, Department of Psychology, Philadelphia, PA, USA
| | - Lauren Zimmermann
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | - Nickilou Y. Krigbaum
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | - Barbara A. Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | | | - Lauren M. Ellman
- Temple University, Department of Psychology, Philadelphia, PA, USA,Corresponding author. Lauren M. Ellman, Ph.D., Temple University, Department of Psychology, Weiss Hall, 1701 North 13 Street, Philadelphia, PA, 19122,
| |
Collapse
|
26
|
Jafari Z, Faraji J, Mirza Agha B, Metz GAS, Kolb BE, Mohajerani MH. The Adverse Effects of Auditory Stress on Mouse Uterus Receptivity and Behaviour. Sci Rep 2017; 7:4720. [PMID: 28680154 PMCID: PMC5498668 DOI: 10.1038/s41598-017-04943-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
Stress during gestation has harmful effects on pregnancy outcome and can lead to spontaneous abortion. Few studies, however, have addressed the impact of gestational stress, particularly auditory stress, on behavioural performance and pregnancy outcome in mice. This study aimed to examine the effect of two types of gestational stress on uterus receptivity and behavioural performance. Pregnant C57BL/6 mice were randomly assigned to either auditory or physical stress conditions or a control condition from gestational days 12-16. The auditory stress regimen used loud 3000 Hz tone, while the physical stressor consisted of restraint and exposure to an elevated platform. Three behavioural tests were performed in the dams after weaning. Uterine receptivity was investigated by counting the number of sites of implantation and fetal resorption. Also, the offspring survival rates during the early postnatal period were calculated. Auditory stress caused an increase in anxiety-like behaviour, reduced time spent exploring new object/environment, and reduced balance when compared to the physical stress and control groups. Auditory stress also caused higher rates of resorbed embryos and reduction of litter size. Our results suggest that the adverse effect of noise stress is stronger than physical stress for both uterus receptivity and behavioural performance of the dams.
Collapse
Affiliation(s)
- Zahra Jafari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
- School of Rehabilitation Sciences, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Jamshid Faraji
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
- Golestan University of Medical Sciences, Faculty of Nursing & Midwifery, Gorgan, Iran
| | - Behroo Mirza Agha
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Gerlinde A S Metz
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| |
Collapse
|
27
|
Abstract
Women undergo developmental and cyclic changes in hormonal exposures that affect brain function and mental health. Some women are more vulnerable to the effects of these hormonal exposures, for reasons that remain to be determined. Evidence to date indicates that anxiety and mood disorders are the most sensitive to hormonal fluctuations in women but there is also growing evidence for a protective effect of female reproductive hormones on schizophrenia. The hormonal exposures of the menstrual cycle, pregnancy, the postpartum period, lactation, and menopause are quite different and may be associated with at least partially distinct symptom profiles.
Collapse
Affiliation(s)
- Margaret Altemus
- VA Connecticut Health Care System, Women's Clinic, Building 2, Room 7-165, 950 Campbell Avenue, New Haven, CT 06516, USA.
| |
Collapse
|
28
|
Effects of combined IUGR and prenatal stress on the development of the hippocampus in a fetal guinea pig model. J Dev Orig Health Dis 2017; 8:584-596. [PMID: 28502262 DOI: 10.1017/s2040174417000307] [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] [Indexed: 11/07/2022]
Abstract
Intrauterine growth restriction (IUGR) and maternal stress during pregnancy are two compromises that negatively impact neurodevelopment and increase the risk of developing later life neuropsychiatric disorders such as schizophrenia, depression and behavioural disorders. Neurosteroids, particularly allopregnanolone, are important in protecting the developing brain and promoting many essential neurodevelopmental processes. Individually, IUGR and prenatal stress (PS) reduce myelination and neurogenesis within affected fetal brains, however less information is available on the combined effects of these two disorders on the term fetal brain. This study aimed to investigate how IUGR and PS impairs the neurosteroid pathway when combined using a guinea pig model, and how these then disrupt the neurodevelopment of the fetus. Uterine artery blood flow restriction was performed at GA30-35 to induce growth restriction, whilst PS was induced by exposure of the dam to a strobe light during gestation commencing GA40 and repeated every 5 days. Exposure in this model caused reductions in hippocampal CA1 MBP immunostaining of male fetuses in both IUGR alone and IUGR+PS paradigms but only by IUGR in the subcortical white mater, compared with control males. Plasma allopregnanolone was reduced by both stressors irrespective of sex, whereas GFAP or MAP2 expression were not affected by either stressor. Female neurodevelopment, as assessed by these markers, was unimpeded by these compromises. The addition of prenatal stress did not further compound these deficits.
Collapse
|
29
|
Ansen-Wilson LJ, Lipinski RJ. Gene-environment interactions in cortical interneuron development and dysfunction: A review of preclinical studies. Neurotoxicology 2017; 58:120-129. [PMID: 27932026 PMCID: PMC5328258 DOI: 10.1016/j.neuro.2016.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/03/2016] [Accepted: 12/03/2016] [Indexed: 12/26/2022]
Abstract
Cortical interneurons (cINs) are a diverse group of locally projecting neurons essential to the organization and regulation of neural networks. Though they comprise only ∼20% of neurons in the neocortex, their dynamic modulation of cortical activity is requisite for normal cognition and underlies multiple aspects of learning and memory. While displaying significant morphological, molecular, and electrophysiological variability, cINs collectively function to maintain the excitatory-inhibitory balance in the cortex by dampening hyperexcitability and synchronizing activity of projection neurons, primarily through use of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Disruption of the excitatory-inhibitory balance is a common pathophysiological feature of multiple seizure and neuropsychiatric disorders, including epilepsy, schizophrenia, and autism. While most studies have focused on genetic disruption of cIN development in these conditions, emerging evidence indicates that cIN development is exquisitely sensitive to teratogenic disruption. Here, we review key aspects of cIN development, including specification, migration, and integration into neural circuits. Additionally, we examine the mechanisms by which prenatal exposure to common chemical and environmental agents disrupt these events in preclinical models. Understanding how genetic and environmental factors interact to disrupt cIN development and function has tremendous potential to advance prevention and treatment of prevalent seizure and neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Lydia J Ansen-Wilson
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Robert J Lipinski
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1010B McArdle Building, 1400 University Avenue, Madison, WI, 53706, USA.
| |
Collapse
|
30
|
Yates NJ, Robertson D, Rodger J, Martin-Iverson MT. Effects of Neonatal Dexamethasone Exposure on Adult Neuropsychiatric Traits in Rats. PLoS One 2016; 11:e0167220. [PMID: 27936175 PMCID: PMC5147874 DOI: 10.1371/journal.pone.0167220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 11/18/2022] Open
Abstract
The effects of early life stress in utero or in neonates has long-term consequences on hypothalamic-pituitary-adrenal (HPA) stress axis function and neurodevelopment. These effects extend into adulthood and may underpin a variety of mental illnesses and be related to various developmental and cognitive changes. We examined the potential role of neonatal HPA axis activation on adult psychopathology and dopamine sensitivity in the mature rat using neonatal exposure to the synthetic glucocorticoid receptor agonist and stress hormone, dexamethasone. We utilized a comprehensive battery of assessments for behaviour, brain function and gene expression to determine if elevated early life HPA activation is associated with adult-onset neuropsychiatric traits. Dexamethasone exposure increased startle reactivity under all conditions tested, but decreased sensitivity of sensorimotor gating to dopaminergic disruption–contrasting with what is observed in several neuropsychiatric diseases. Under certain conditions there also appeared to be mild long-term changes in stress and anxiety-related behaviours with neonatal dexamethasone exposure. Electrophysiology revealed that there were no consistent neuropsychiatric abnormalities in auditory processing or resting state brain function with dexamethasone exposure. However, neonatal dexamethasone altered auditory cortex glucocorticoid activation, and auditory cortex synchronization. Our results indicate that neonatal HPA axis activation by dexamethasone alters several aspects of adult brain function and behaviour and may induce long-term changes in emotional stress-reactivity. However, neonatal dexamethasone exposure is not specifically related to any particular neuropsychiatric disease.
Collapse
Affiliation(s)
- Nathanael J. Yates
- School of Animal Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
- * E-mail:
| | - Donald Robertson
- School of Anatomy, Physiology, and Human Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Jennifer Rodger
- School of Animal Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Mathew T. Martin-Iverson
- School of Medicine and Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
31
|
Kalinina DS, Vol’nova AB, Alekseeva OS, Zhuravin IA. Electrical activity of the neocortex in adult rats after prenatal hypoxia and in epilepsy model. J EVOL BIOCHEM PHYS+ 2016. [DOI: 10.1134/s0022093016050033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Geary DC. Evolution of Sex Differences in Trait- and Age-Specific Vulnerabilities. PERSPECTIVES ON PSYCHOLOGICAL SCIENCE 2016; 11:855-876. [DOI: 10.1177/1745691616650677] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Traits that facilitate competition for reproductive resources or that influence mate choice generally have a heightened sensitivity to stressors. They have evolved to signal resilience to infectious disease and nutritional and social stressors, and they are compromised by exposure to man-made toxins. Although these traits can differ from one species or sex to the next, an understanding of the dynamics of competition and choice can in theory be used to generate a priori predictions about sex-, age-, and trait-specific vulnerabilities for any sexually reproducing species. I provide a review of these dynamics and illustrate associated vulnerabilities in nonhuman species. The age- and sex-specific vulnerability of such traits is then illustrated for stressor-related disruptions of boys’ and girls’ physical growth and play behavior, as well as for aspects of boys’ and girls’ and men’s and women’s personality, language, and spatial abilities. There is much that remains to be determined, but enough is now known to reframe trait sensitivity in ways that will allow scientists and practitioners to better identify and understand vulnerable human traits, and eventually ameliorate or prevent their expression.
Collapse
|
33
|
Faa G, Manchia M, Pintus R, Gerosa C, Marcialis MA, Fanos V. Fetal programming of neuropsychiatric disorders. ACTA ACUST UNITED AC 2016; 108:207-223. [DOI: 10.1002/bdrc.21139] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Gavino Faa
- Division of Pathology, Department of Surgery; University Hospital San Giovanni di Dio; Cagliari Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Public Health, Clinical and Molecular Medicine; University of Cagliari; Cagliari Italy
- Department of Pharmacology; Dalhousie University; Halifax Nova Scotia Canada
| | - Roberta Pintus
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| | - Clara Gerosa
- Division of Pathology, Department of Surgery; University Hospital San Giovanni di Dio; Cagliari Italy
| | - Maria Antonietta Marcialis
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| |
Collapse
|
34
|
Epigenetic legacy of parental experiences: Dynamic and interactive pathways to inheritance. Dev Psychopathol 2016; 28:1219-1228. [PMID: 27687718 DOI: 10.1017/s0954579416000808] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The quality of the environment experienced by an individual across his or her lifespan can result in a unique developmental trajectory with consequences for adult phenotype and reproductive success. However, it is also evident that these experiences can impact the development of offspring with continued effect on subsequent generations. Epigenetic mechanisms have been proposed as a mediator of both these within- and across-generation effects, and there is increasing evidence to support the role of environmentally induced changes in DNA methylation, posttranslational histone modifications, and noncoding RNAs in predicting these outcomes. Advances in our understanding of these molecular modifications contribute to increasingly nuanced perspectives on plasticity and transmission of phenotypes across generations. A challenge that emerges from this research is in how we integrate these "new" perspectives with traditional views of development, reproduction, and inheritance. This paper will highlight evidence suggestive of an epigenetic impact of the environment on mothers, fathers, and their offspring, and illustrate the importance of considering the dynamic nature of reproduction and development and inclusive views of inheritance within the evolving field of behavioral and environmental epigenetics.
Collapse
|
35
|
Montes P, Ruiz-Sánchez E, Calvillo M, Rojas P. Active coping of prenatally stressed rats in the forced swimming test: involvement of the Nurr1 gene. Stress 2016; 19:506-15. [PMID: 27219004 DOI: 10.1080/10253890.2016.1193147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Depending on genetic predisposition, prenatal stress may result in vulnerability or resilience to develop psychiatric disorders in adulthood. Nurr1 is an immediate early gene, important in the brain for the stress response. We tested the hypothesis that prenatal stress and the decrease of hippocampal Nurr1 alter offspring behavioral responses in the forced swimming test (FST). Pregnant Wistar rats were exposed to restraint stress (45 min, thrice daily) from gestation day 14. Prenatally stressed (PS) and non-prenatally stressed (NPS) male offspring were treated bilaterally with a Nurr1 antisense oligodeoxynucleotide (ODN; or control) into the hippocampus at 97 d of age. After 1 h, the rats were exposed to the FST (acute stressor) to analyze their behavioral responses. Thirty minutes after the FST, we analyzed the gene expression of Nurr1, Bdnf and Nr3c1 (genes for Nurr1, brain-derived neurotrophic factor (BDNF) and glucocorticoid receptor (GR), respectively) in the hippocampus, prefrontal cortex (PFC) and hypothalamus. Results showed that the decrease of hippocampal Nurr1 after the antisense ODN in adult NPS rats induces immobility (indicating depressive-like behavior). The PS adult rats, including the group with decreased hippocampal Nurr1, presented low immobility in the FST. This low immobility was concordant with maintenance of Nurr1 and Bdnf expression levels in the three analyzed brain regions; Nr3c1 gene expression was also maintained in the PFC and hypothalamus. These findings suggest that Nurr1 and associated genes could participate in the brain modifications induced by prenatal stress, allowing active coping (resilience) with acute stress in adulthood.
Collapse
MESH Headings
- Adaptation, Psychological/physiology
- Animals
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Female
- Gene Expression
- Hippocampus/metabolism
- Hypothalamus/metabolism
- Male
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Prefrontal Cortex/metabolism
- Pregnancy
- Prenatal Exposure Delayed Effects/genetics
- Prenatal Exposure Delayed Effects/metabolism
- Prenatal Exposure Delayed Effects/psychology
- Rats
- Rats, Wistar
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Restraint, Physical
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/psychology
- Swimming/psychology
Collapse
Affiliation(s)
- Pedro Montes
- a Laboratory of Neurotoxicology , National Institute of Neurology and Neurosurgery, "Manuel Velasco Suárez" , Mexico D.F. , Mexico
| | - Elizabeth Ruiz-Sánchez
- a Laboratory of Neurotoxicology , National Institute of Neurology and Neurosurgery, "Manuel Velasco Suárez" , Mexico D.F. , Mexico
| | - Minerva Calvillo
- b Experimental Laboratory of Neurodegenerative Diseases , National Institute of Neurology and Neurosurgery, "Manuel Velasco Suárez" , Mexico D.F. , Mexico
| | - Patricia Rojas
- a Laboratory of Neurotoxicology , National Institute of Neurology and Neurosurgery, "Manuel Velasco Suárez" , Mexico D.F. , Mexico
| |
Collapse
|
36
|
Hiroi R, Carbone DL, Zuloaga DG, Bimonte-Nelson HA, Handa RJ. Sex-dependent programming effects of prenatal glucocorticoid treatment on the developing serotonin system and stress-related behaviors in adulthood. Neuroscience 2016; 320:43-56. [PMID: 26844389 PMCID: PMC4840233 DOI: 10.1016/j.neuroscience.2016.01.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/12/2016] [Accepted: 01/26/2016] [Indexed: 12/25/2022]
Abstract
Prenatal stress and overexposure to glucocorticoids (GC) during development may be associated with an increased susceptibility to a number of diseases in adulthood including neuropsychiatric disorders, such as depression and anxiety. In animal models, prenatal overexposure to GC results in hyper-responsiveness to stress in adulthood, and females appear to be more susceptible than males. Here, we tested the hypothesis that overexposure to GC during fetal development has sex-specific programming effects on the brain, resulting in altered behaviors in adulthood. We examined the effects of dexamethasone (DEX; a synthetic GC) during prenatal life on stress-related behaviors in adulthood and on the tryptophan hydroxylase-2 (TpH2) gene expression in the adult dorsal raphe nucleus (DRN). TpH2 is the rate-limiting enzyme for serotonin (5-HT) synthesis and has been implicated in the etiology of human affective disorders. Timed-pregnant rats were treated with DEX from gestational days 18-22. Male and female offspring were sacrificed on the day of birth (postnatal day 0; P0), P7, and in adulthood (P80-84) and brains were examined for changes in TpH2 mRNA expression. Adult animals were also tested for anxiety- and depressive- like behaviors. In adulthood, prenatal DEX increased anxiety- and depressive- like behaviors selectively in females, as measured by decreased time spent in the center of the open field and increased time spent immobile in the forced swim test, respectively. Prenatal DEX increased TpH2 mRNA selectively in the female caudal DRN at P7, whereas it decreased TpH2 mRNA selectively in the female caudal DRN in adulthood. In animals challenged with restraint stress in adulthood, TpH2 mRNA was significantly lower in rostral DRN of prenatal DEX-treated females compared to vehicle-treated females. These data demonstrated that prenatal overexposure to GC alters the development of TpH2 gene expression and these alterations correlated with lasting behavioral changes found in adult female offspring.
Collapse
Affiliation(s)
- R Hiroi
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA; Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - D L Carbone
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - D G Zuloaga
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - H A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - R J Handa
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| |
Collapse
|
37
|
Fineberg AM, Ellman LM, Schaefer CA, Maxwell SD, Shen L, Chaudhury NH, Cook AL, Bresnahan MA, Susser ES, Brown AS. Fetal exposure to maternal stress and risk for schizophrenia spectrum disorders among offspring: Differential influences of fetal sex. Psychiatry Res 2016; 236:91-97. [PMID: 26753951 PMCID: PMC4767153 DOI: 10.1016/j.psychres.2015.12.026] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/15/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023]
Abstract
Exposure to adverse life events during pregnancy has been linked to increased risk of schizophrenia spectrum disorders (SSD) in offspring. Nevertheless, much of the previous work inferred maternal stress from severe life events rather than directly assessing maternal reports of stress. The present study aimed to examine maternal reports of stress during pregnancy and risk for offspring SSD. Participants were 95 SSD cases and 206 controls who were offspring from a large birth cohort study that followed pregnant women from 1959 to 1966. During pregnancy interviews, women were asked if anything worrisome had occurred recently. Interviews were qualitatively coded for stress-related themes, including reports of daily life stress, by two independent raters. None of the maternal psychosocial stress themes were significantly associated with increased odds of offspring SSD in analyses of the full sample. However, results indicated a significant daily life stress by infant sex interaction. Maternal daily life stress during pregnancy was associated with significantly increased odds of SSD among male offspring. Findings suggest sex-specific fetal sensitivity to maternal reported daily life stress during pregnancy on risk for SSD, with males appearing to be more vulnerable to the influences of maternal stress during pregnancy.
Collapse
Affiliation(s)
- Anna M Fineberg
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, Philadelphia, PA, USA.
| | | | - Seth D Maxwell
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Ling Shen
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | - Nashid H Chaudhury
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA
| | - Aundrea L Cook
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Michaeline A Bresnahan
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Ezra S Susser
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Alan S Brown
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
38
|
Bowers ME, Yehuda R. Intergenerational Transmission of Stress in Humans. Neuropsychopharmacology 2016; 41:232-44. [PMID: 26279078 PMCID: PMC4677138 DOI: 10.1038/npp.2015.247] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/19/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023]
Abstract
The hypothesis that offspring are affected by parental trauma or stress exposure, first noted anecdotally, is now supported empirically by data from Holocaust survivor offspring cohorts and other populations. These findings have been extended to less extreme forms of stress, where differential physical, behavioral, and cognitive outcomes are observed in affected offspring. Parental stress-mediated effects in offspring could be explained by genetics or social learning theory. Alternatively, biological variations stemming from stress exposure in parents could more directly have an impact on offspring, a concept we refer to here as 'intergenerational transmission', via changes to gametes and the gestational uterine environment. We further extend this definition to include the transmission of stress to offspring via early postnatal care, as animal studies demonstrate the importance of early maternal care of pups in affecting offsprings' long-term behavioral changes. Here, we review clinical observations in offspring, noting that offspring of stress- or trauma-exposed parents may be at greater risk for physical, behavioral, and cognitive problems, as well as psychopathology. Furthermore, we review findings concerning offspring biological correlates of parental stress, in particular, offspring neuroendocrine, epigenetic, and neuroanatomical changes, in an attempt to determine the extent of parental stress effects. Although understanding the etiology of effects in offspring is currently impeded by methodological constraints, and limitations in our knowledge, we summarize current information and conclude by presenting hypotheses that have been prompted by recent studies in the field.
Collapse
Affiliation(s)
- Mallory E Bowers
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Rachel Yehuda
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, NY, USA,Mental Health Care Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA,Department of Neuroscience, Icahn School of Medicine at Mount, NY, NY, USA,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, James J. Peters Veterans Affairs Medical Center, 526 OOMH PTSD 116/A, JJP VAMC, 130 W Kingsbridge Road, Bronx, NY 10468, USA, Tel: +718 741 4000, ext. 6964, Fax: +718 741 4703, E-mail:
| |
Collapse
|
39
|
Flinkkilä E, Keski-Rahkonen A, Marttunen M, Raevuori A. Prenatal Inflammation, Infections and Mental Disorders. Psychopathology 2016; 49:317-333. [PMID: 27529630 DOI: 10.1159/000448054] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The objective of this descriptive review is to summarize the current scientific evidence on the effect of prenatal exposure to maternal infection and immune response on the offspring's risk for mental disorders (schizophrenia spectrum disorders, autism spectrum disorders, attention-deficit hyperactivity disorder, anorexia nervosa, and mood disorders). SAMPLING AND METHODS Studies were searched from PubMed and Ovid MEDLINE (R) databases with the following keywords: 'prenatal exposure delayed effects' and 'infection', and 'inflammation' and 'mental disorders'. A comprehensive manual search, including a search from the reference list of included articles, was also performed. RESULTS Prenatal exposure to maternal influenza appears to increase the offspring's risk for schizophrenia spectrum disorders, although studies are not fully consistent. Prenatal exposure to maternal fever and elevated cytokine levels seems to be related to the elevated risk for autism spectrum disorders in the offspring. No replicated findings of an association between prenatal infectious exposure and other mental disorders exist. CONCLUSIONS Evidence for the effect of prenatal exposure to maternal infection on risk for mental disorders exists for several different infections, suggesting that common factors occurring in infections (e.g. elevated cytokine levels and fever), rather than the infectious agent itself, might be the underlying factor in increasing the risk for mental disorders. Additionally, it is likely that genetic liability to these disorders operates in conjunction with the exposure. Therefore, genetically sensitive study designs are needed in future studies.
Collapse
Affiliation(s)
- Eerika Flinkkilä
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
40
|
Cowan CSM, Callaghan BL, Kan JM, Richardson R. The lasting impact of early-life adversity on individuals and their descendants: potential mechanisms and hope for intervention. GENES BRAIN AND BEHAVIOR 2015; 15:155-68. [PMID: 26482536 DOI: 10.1111/gbb.12263] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 01/15/2023]
Abstract
The adverse effects of early-life stress are pervasive, with well-established mental and physical health consequences for exposed individuals. The impact of early adverse experiences is also highly persistent, with documented increases in risk for mental illness across the life span that are accompanied by stable alterations in neural function and hormonal responses to stress. Here, we review some of these 'stress phenotypes', with a focus on intermediary factors that may signal risk for long-term mental health outcomes, such as altered development of the fear regulation system. Intriguingly, recent research suggests that such stress phenotypes may persist even beyond the life span of the individuals, with consequences for their offspring and grand-offspring. Phenotypic characteristics may be transmitted to future generations via either the matriline or the patriline, a phenomenon that has been demonstrated in both human and animal studies. In this review, we highlight behavioral and epigenetic factors that may contribute to this multigenerational transmission and discuss the potential of various treatment approaches that may halt the cycle of stress phenotypes.
Collapse
Affiliation(s)
- C S M Cowan
- School of Psychology, The University of New South Wales, Sydney, Australia
| | - B L Callaghan
- Psychology Department, Columbia University, New York, NY, USA
| | - J M Kan
- School of Psychology, The University of New South Wales, Sydney, Australia
| | - R Richardson
- School of Psychology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
41
|
Association Between Prenatal Exposure to Maternal Infection and Offspring Mood Disorders: A Review of the Literature. Curr Probl Pediatr Adolesc Health Care 2015; 45:325-64. [PMID: 26476880 DOI: 10.1016/j.cppeds.2015.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/14/2015] [Indexed: 01/19/2023]
Abstract
The purpose of this article is to provide a systematic review of studies that have examined the association between prenatal exposure to maternal infection and development of mood disorders across the life course. Drawing from both human- and animal-based studies, we give an overview of hypothesized biological mechanisms by which exposure to maternal infection during critical periods of gestation may contribute to fetal programming of mood disorders in offspring. We discuss studies examining the association between prenatal exposure to maternal infection with pathogens including influenza as well as other respiratory viruses, herpesviruses, hepatitis viruses, and Toxoplasma gondii and mood disorders in human populations. Moreover, we outline strengths and limitations of the current body of evidence and make recommendations for future research. We also discuss findings in the context of well-documented gender and socioeconomic disparities in the prevalence and severity of mood disorders, particularly major depression, and the role that early exposure to infection may play in explaining the perpetuation of such disparities across generations. Overall, this review of the current knowledge on this topic has important implications for determining future research directions, designing interventions as well as prenatal care guidelines targeted at prevention or treatment of infection during pregnancy, and clinical practice for the identification of individuals that may be at increased risk for mood disorders beginning early in life. Importantly, such efforts may not only lower the overall burden of mood disorders but also serve to address social disparities in these adverse mental health conditions in the U.S.
Collapse
|
42
|
Bennett GA, Palliser HK, Shaw JC, Walker D, Hirst JJ. Prenatal Stress Alters Hippocampal Neuroglia and Increases Anxiety in Childhood. Dev Neurosci 2015; 37:533-45. [DOI: 10.1159/000437302] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/30/2015] [Indexed: 11/19/2022] Open
Abstract
Prenatal stress has been associated with detrimental outcomes of pregnancy, including altered brain development leading to behavioural pathologies. The neurosteroid allopregnanolone has been implicated in mediating some of these adverse outcomes following prenatal stress due to its potent inhibitory and anxiolytic effects on the brain. The aims of the current study were to characterise key markers for brain development as well as behavioural parameters, adrenocortical responses to handling and possible neurosteroid influences towards outcomes in guinea pig offspring in childhood. Pregnant guinea pig dams were exposed to strobe light for 2 h (9-11 a.m.) on gestational days 50, 55, 60, and 65 and were left to deliver spontaneously at term and care for their litter. Behavioural testing (open-field test, object exploration test) of the offspring was performed at postnatal day 18 (with salivary cortisol and DHEA measured), and brains were collected at post-mortem on day 21. Markers of brain development myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) were assessed via immunohistochemistry, and the neurosteroid allopregnanolone and its rate-limiting enzymes 5α-reductase types 1 and 2 (5αR1/2) were measured in neonatal brains by radioimmunoassay, reverse transcriptase polymerase chain reaction (RT-PCR), and Western blot, respectively. Brain-derived neurotrophic factor protein was measured as a marker of synaptic plasticity, and GABAA receptor subunit expression was also assessed using RT-PCR. Neonates born from mothers stressed during late pregnancy showed a reduction in both MBP (p < 0.01) and GFAP (p < 0.05) expression in the CA1 region of the hippocampus at 21 days of age. Pups of prenatally stressed pregnancies also showed higher levels of anxiety and neophobic behaviours at the equivalent of childhood (p < 0.05). There were no significant changes observed in allopregnanolone levels, 5αR1/2 expression, or GABAA receptor subunit expression in prenatally stressed neonates compared to controls. This study shows alterations in markers of myelination and reactive astrocytes in the hippocampus of offspring exposed to prenatal stress. These changes are also observed in offspring that show increased anxiety behaviours at the equivalent of childhood, which indicates ongoing structural and functional postnatal changes after prenatal stress exposure.
Collapse
|
43
|
Hoffman K, Aschengrau A, Webster TF, Bartell SM, Vieira VM. Associations between residence at birth and mental health disorders: a spatial analysis of retrospective cohort data. BMC Public Health 2015; 15:688. [PMID: 26195105 PMCID: PMC4508761 DOI: 10.1186/s12889-015-2011-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mental health disorders impact approximately one in four US adults. While their causes are likely multifactorial, prior research has linked the risk of certain mental health disorders to prenatal and early childhood environmental exposures, motivating a spatial analysis to determine whether risk varies by birth location. METHODS We investigated the spatial associations between residence at birth and odds of depression, bipolar disorder, and post-traumatic stress disorder (PTSD) in a retrospective cohort (Cape Cod, Massachusetts, 1969-1983) using generalized additive models to simultaneously smooth location and adjust for confounders. Birth location served as a surrogate for prenatal exposure to the combination of social and environmental factors related to the development of mental illness. We predicted crude and adjusted odds ratios (aOR) for each outcome across the study area. The results were mapped to identify areas of increased risk. RESULTS We observed spatial variation in the crude odds ratios of depression that was still present even after accounting for spatial confounding due to geographic differences in the distribution of known risk factors (aOR range: 0.61-3.07, P = 0.03). Similar geographic patterns were seen for the crude odds of PTSD; however, these patterns were no longer present in the adjusted analysis (aOR range: 0.49-1.36, P = 0.79), with family history of mental illness most notably influencing the geographic patterns. Analyses of the odds of bipolar disorder did not show any meaningful spatial variation (aOR range: 0.58-1.17, P = 0.82). CONCLUSION Spatial associations exist between residence at birth and odds of PTSD and depression, but much of this variation can be explained by the geographic distributions of available risk factors. However, these risk factors did not account for all the variation observed with depression, suggesting that other social and environmental factors within our study area need further investigation.
Collapse
Affiliation(s)
- Kate Hoffman
- Nicholas School of the Environment, Duke University, Durham, NC, USA.
| | - Ann Aschengrau
- Boston University School of Public Health, Boston, MA, 02118, USA.
| | - Thomas F Webster
- Boston University School of Public Health, Boston, MA, 02118, USA.
| | - Scott M Bartell
- Program in Public Health, University of California, Irvine, 653 E. Peltason Drive, AIRB 2042, Irvine, CA, 92697, USA.
| | - Verónica M Vieira
- Program in Public Health, University of California, Irvine, 653 E. Peltason Drive, AIRB 2042, Irvine, CA, 92697, USA.
| |
Collapse
|
44
|
Alterations of Glucocorticoid Receptor Gene Methylation in Externalizing Disorders During Childhood and Adolescence. Behav Genet 2015; 45:529-36. [DOI: 10.1007/s10519-015-9721-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 04/07/2015] [Indexed: 10/23/2022]
|
45
|
Betts KS, Williams GM, Najman JM, Alati R. The relationship between maternal depressive, anxious, and stress symptoms during pregnancy and adult offspring behavioral and emotional problems. Depress Anxiety 2015; 32:82-90. [PMID: 24788841 DOI: 10.1002/da.22272] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/11/2014] [Accepted: 03/15/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Prenatal maternal depressive, anxious, and stress symptoms have been found to be associated with child and adolescent behavior problems. In this paper, we investigate their impact on behavior problems and depressive symptoms in adulthood. METHODS Participants included 3,099 mother-offspring pairs from the Mater University Study of Pregnancy (MUSP), an Australian based, prebirth cohort study. We used latent class growth analysis (LCGA) with parallel processes to identify trajectories of maternal depressive, anxious, and stress symptoms over four time periods between the mothers' first clinic visit and 5 years postpregnancy. We fitted the estimates from the maternal trajectories in multivariate logistic regression models to predict internalizing and externalizing behavior at age 21. We adjusted for a wide range of prenatal and postnatal factors, including maternal life events, relationship quality, contact with the new born, as well as concurrent maternal depressive and anxious symptoms and father's history of mental health problem. RESULTS LCGA found seven groups of mothers; one group of mothers exhibited high levels of depressive, anxious, and stress symptoms during pregnancy but not at later time points. Their offspring experienced increased levels of behavior problems and depressive symptoms. CONCLUSIONS This paper provides the first evidence that high levels of maternal subjective depressive, anxious, and stress symptoms experienced in early pregnancy may predict internalizing and externalizing behavior problems and depressive symptoms in young adults.
Collapse
Affiliation(s)
- Kim S Betts
- School of Population Health, University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
46
|
Skelin I, Needham MA, Molina LM, Metz GAS, Gruber AJ. Multigenerational prenatal stress increases the coherence of brain signaling among cortico-striatal-limbic circuits in adult rats. Neuroscience 2015; 289:270-8. [PMID: 25595989 DOI: 10.1016/j.neuroscience.2015.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/23/2014] [Accepted: 01/04/2015] [Indexed: 11/28/2022]
Abstract
Prenatal stress (PNS) is a significant risk factor for the development of psychopathology in adulthood such as anxiety, depression, schizophrenia and addiction. Animal models of PNS resemble many of the effects of PNS on humans and provide a means to study the accumulated effects of PNS over several generations on brain function. Here, we examined how mild PNS delivered during the third week in utero over four consecutive generations affects behavioral flexibility and functional signaling among cortical and limbic structures. These multi-generational prenatally stressed (MGPNS) rats were not impaired on an odor-cued reversal learning task as compared to control animals. Unilateral field potential (FP) recordings from the medial prefrontal cortex, basolateral amygdala, ventral hippocampus, and striatal territories revealed widespread differences in brain signaling between these groups during the odor sampling phase of the task. The FP power was significantly lower in most structures across most frequency bands in MGPNS animals, and the relative increase in power from baseline during the task was lower for the beta band (12-30Hz) in MGPNS animals as compared to controls. The coherence of FPs between brain regions, however, was much higher in MGPNS animals among all structures and for most frequency bands. We propose that this pattern of changes in brain signaling reflects a simplification of network processing, which is consistent with reports of reduced spine density and dendritic complexity in the brains of animals receiving PNS. Our data support the proposal that recurrent ancestral stress leads to adaptations in the brain, and that these may confer adaptive behavior in some circumstances as compared to single-generation PNS.
Collapse
Affiliation(s)
- I Skelin
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - M A Needham
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - L M Molina
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - G A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - A J Gruber
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.
| |
Collapse
|
47
|
Schoenfeld TJ, Cameron HA. Adult neurogenesis and mental illness. Neuropsychopharmacology 2015; 40:113-28. [PMID: 25178407 PMCID: PMC4262910 DOI: 10.1038/npp.2014.230] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023]
Abstract
Several lines of evidence suggest that adult neurogenesis, the production of new neurons in adulthood, may play a role in psychiatric disorders, including depression, anxiety, and schizophrenia. Medications and other treatments for mental disorders often promote the proliferation of new neurons; the time course for maturation and integration of new neurons in circuitry parallels the delayed efficacy of psychiatric therapies; adverse and beneficial experiences similarly affect development of mental illness and neurogenesis; and ablation of new neurons in adulthood alters the behavioral impact of drugs in animal models. At present, the links between adult neurogenesis and depression seem stronger than those suggesting a relationship between new neurons and anxiety or schizophrenia. Yet, even in the case of depression there is currently no direct evidence for a causative role. This article reviews the data relating adult neurogenesis to mental illness and discusses where research needs to head in the future.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA,Section on Neuroplasticity, NIMH, 35 Convent Drive, Building 35/3C915, Bethesda, MD 20892-3718, USA, Tel: +1 301 496 3814, Fax: +1 301 480 4564, E-mail:
| |
Collapse
|
48
|
Abstract
ABSTRACT
Fetal development and growth, as well as the timing of birth is influenced by the intrauterine environment. Many environmental factors causing the fetal stress can interfere with fetal development and leave long-term and profound consequences on health. Fetal glucocorticoid overexposure has primarily significant consequences for the development of the central nervous system. In response to an adverse intrauterine conditions, the fetus is able to adapt its physiology to promote survival. However, these adaptations can result in permanent changes in tissue and organ structure and function that directly ‘program’ predisposition to disease. Cardiometabolic disorders, behavioral alterations and neuropsychiatric impairments in adulthood and/ or childhood may have their roots in the fetal period of life. Fetal response to stress and its prenatal and lifelong consequences are discussed in this review.
How to cite this article
Kadić AS. Fetal Neurology: The Role of Fetal Stress. Donald School J Ultrasound Obstet Gynecol 2015;9(1):30-39.
Collapse
|
49
|
Boersma GJ, Tamashiro KL. Individual differences in the effects of prenatal stress exposure in rodents. Neurobiol Stress 2015; 1:100-8. [PMID: 27589662 PMCID: PMC4721332 DOI: 10.1016/j.ynstr.2014.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/18/2023] Open
Abstract
Exposure to prenatal stress alters the phenotype of the offspring in adulthood. When the prenatal and adult environments do not match, these alterations may induce pathology risk. There are, however, large individual differences in the effects of prenatal stress. While some individuals seem vulnerable, others appear to be relatively resistant to its effects. In this review we discuss potential mechanisms underlying these individual differences with a focus on animal models. Differences between rodent models selected for stress coping traits are discussed. In addition, the role of circulating factors, like glucocorticoids and cytokines, factors involved in brain development and influences of epigenetic and genetic factors in prenatal stress induced phenotype are covered.
Collapse
Affiliation(s)
- Gretha J. Boersma
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
50
|
Babenko O, Kovalchuk I, Metz GAS. Stress-induced perinatal and transgenerational epigenetic programming of brain development and mental health. Neurosci Biobehav Rev 2014; 48:70-91. [PMID: 25464029 DOI: 10.1016/j.neubiorev.2014.11.013] [Citation(s) in RCA: 335] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/19/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
Research efforts during the past decades have provided intriguing evidence suggesting that stressful experiences during pregnancy exert long-term consequences on the future mental wellbeing of both the mother and her baby. Recent human epidemiological and animal studies indicate that stressful experiences in utero or during early life may increase the risk of neurological and psychiatric disorders, arguably via altered epigenetic regulation. Epigenetic mechanisms, such as miRNA expression, DNA methylation, and histone modifications are prone to changes in response to stressful experiences and hostile environmental factors. Altered epigenetic regulation may potentially influence fetal endocrine programming and brain development across several generations. Only recently, however, more attention has been paid to possible transgenerational effects of stress. In this review we discuss the evidence of transgenerational epigenetic inheritance of stress exposure in human studies and animal models. We highlight the complex interplay between prenatal stress exposure, associated changes in miRNA expression and DNA methylation in placenta and brain and possible links to greater risks of schizophrenia, attention deficit hyperactivity disorder, autism, anxiety- or depression-related disorders later in life. Based on existing evidence, we propose that prenatal stress, through the generation of epigenetic alterations, becomes one of the most powerful influences on mental health in later life. The consideration of ancestral and prenatal stress effects on lifetime health trajectories is critical for improving strategies that support healthy development and successful aging.
Collapse
Affiliation(s)
- Olena Babenko
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4; Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| |
Collapse
|