101
|
O'Connor T, Best M, Brunner J, Ciesla AA, Cunning A, Kapula N, Kautz A, Khoury L, Macomber A, Meng Y, Miller RK, Murphy H, Salafia CM, Vallejo Sefair A, Serrano J, Barrett E. Cohort profile: Understanding Pregnancy Signals and Infant Development (UPSIDE): a pregnancy cohort study on prenatal exposure mechanisms for child health. BMJ Open 2021; 11:e044798. [PMID: 33795306 PMCID: PMC8021752 DOI: 10.1136/bmjopen-2020-044798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 01/28/2023] Open
Abstract
PURPOSE Extensive research suggests that maternal prenatal distress is reliably related to perinatal and child health outcomes-which may persist into adulthood. However, basic questions remain regarding mechanisms involved. To better understand these mechanisms, we developed the Understanding Pregnancy Signals and Infant Development (UPSIDE) cohort study, which has several distinguishing features, including repeated assessments across trimesters, analysis of multiple biological pathways of interest, and incorporation of placental structure and function as mediators of child health outcomes. PARTICIPANTS Women with normal risk pregnancies were recruited at <14 weeks gestation. Study visits occurred in each trimester and included extensive psychological, sociodemographic, health behaviour and biospecimen collection. Placenta and cord blood were collected at birth. Child visits (ongoing) occur at birth and 1, 6, 12, 24, 36 and 48 months of age and use standard anthropometric, clinical, behavioural, biological and neuroimaging methods to assess child physical and neurodevelopment. FINDINGS TO DATE We recruited 326 pregnancies; 294 (90%) were retained through birth. Success rates for prenatal biospecimen collection were high across all trimesters (96%-99% for blood, 94%-97% for urine, 96%-99% for saliva, 96% of placentas, 88% for cord blood and 93% for buccal swab). Ninety-four per cent of eligible babies (n=277) participated in a birth examination; postnatal visits are ongoing. FUTURE PLANS The current phase of the study follows children through age 4 to examine child neurodevelopment and physical development. In addition, the cohort participates in the National Institutes of Health's Environmental influences on Child Health Outcomes programme, a national study of 50 000 families examining early environmental influences on perinatal outcomes, neurodevelopment, obesity and airway disease. Future research will leverage the rich repository of biological samples and clinical data to expand research on the mechanisms of child health outcomes in relation to environmental chemical exposures, genetics and the microbiome.
Collapse
Affiliation(s)
- Thomas O'Connor
- Psychiatry, University of Rochester, Rochester, New York, USA
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
- Neuroscience, University of Rochester Medical Center, Rochester, New York, USA
- Wynne Center for Family Research, University of Rochester Medical Center, Rochester, New York, USA
- Psychology, University of Rochester, Rochester, New York, USA
| | - Meghan Best
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jessica Brunner
- Psychiatry, University of Rochester, Rochester, New York, USA
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
- School of Nursing, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Allison Cunning
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ntemena Kapula
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
- School of Nursing, University of Rochester Medical Center, Rochester, New York, USA
| | - Amber Kautz
- Public Health Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Leena Khoury
- Psychiatry, University of Rochester, Rochester, New York, USA
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Allison Macomber
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ying Meng
- School of Nursing, University of Rochester Medical Center, Rochester, New York, USA
| | - Richard K Miller
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Hannah Murphy
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Carolyn M Salafia
- Placental Modulation Laboratory, Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
- Placental Analytics LLC, Larchmont, New York, USA
| | | | - Jishyra Serrano
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Emily Barrett
- Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
- Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
102
|
Berry A, Mazzelli M, Musillo C, Riva MA, Cattaneo A, Cirulli F. High-fat diet during adulthood interacts with prenatal stress, affecting both brain inflammatory and neuroendocrine markers in male rats. Eur J Neurosci 2021; 55:2326-2340. [PMID: 33711185 DOI: 10.1111/ejn.15181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023]
Abstract
Prenatal stress (PNS) affects foetal programming and, through an interaction with subsequent challenges, can increase vulnerability to mood and metabolic disorders. We have previously shown that, following PNS, adult male rats are characterized by increased vulnerability to a metabolic stressor experienced at adulthood (8-week-high-fat diet-HFD). In this study, we specifically assessed whether PNS might interact with an adult metabolic challenge to induce an inflammatory phenotype. Changes in the expression levels of inflammatory (Il-1β, Tnf-α, Il-6) and of stress response mediators (Nr3c1, Fkbp5) as well as of mood and metabolic regulators (Bdnf, Ghs-R) were investigated in the hippocampus, prefrontal cortex and hypothalamus, brain regions involved in the pathogenesis of depression and prone to inflammation in response to stress. Overall, PNS reduced the expression of Bdnf and Tnf-α, while HFD administered at adulthood counteracted this effect suggesting that PNS impinges upon the same pathways regulating responses to a metabolic challenge at adulthood. Furthermore, HFD and PNS affected the expression of both Nr3c1 and Fkbp5, two neuroendocrine mediators involved in the response to stress, metabolic challenges and in the modulation of the emotional profile (as shown by the correlation between Fkbp5 and the time spent in the open arms of the elevated plus-maze). Overall, these results indicate that the same metabolic and neuroendocrine effectors engaged by PNS are affected by metabolic challenges at adulthood, providing some mechanistic insight into the well-known comorbidity between mood and metabolic disorders.
Collapse
Affiliation(s)
- Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Mazzelli
- Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.,PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Marco Andrea Riva
- Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
103
|
Associations between Dietary Patterns and Inflammatory Markers during Pregnancy: A Systematic Review. Nutrients 2021; 13:nu13030834. [PMID: 33806342 PMCID: PMC8000934 DOI: 10.3390/nu13030834] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Elevated inflammation in pregnancy has been associated with multiple adverse pregnancy outcomes and potentially an increased susceptibility to future chronic disease. How maternal dietary patterns influence systemic inflammation during pregnancy requires further investigation. The purpose of this review was to comprehensively evaluate studies that assessed dietary patterns and inflammatory markers during pregnancy. This review was guided by the Preferred Reporting Items for Systematic Review and Meta-Analyses. Included studies were sourced from EMBASE, PubMed, Web of Science, and Scopus and evaluated using The Quality Assessment Tool for Quantitative Studies. Inclusion criteria consisted of human studies published in English between January 2007 and May 2020 that addressed associations between dietary patterns and inflammatory markers during pregnancy. Studies focused on a single nutrient, supplementation, or combined interventions were excluded. A total of 17 studies were included. Despite some inconsistent findings, maternal diets characterized by a higher intake of animal protein and cholesterol and/or a lower intake of fiber were shown to be associated with certain pro-inflammatory markers (C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor-α (TNF- α), IL-8, serum amyloid A (SAA), and glycoprotein acetylation (GlycA)). Future studies that explore a broader range of inflammatory markers in the pregnant population, reduce measurement errors, and ensure adequate statistical adjustment are warranted.
Collapse
|
104
|
Lu YC, Kapse K, Andersen N, Quistorff J, Lopez C, Fry A, Cheng J, Andescavage N, Wu Y, Espinosa K, Vezina G, du Plessis A, Limperopoulos C. Association Between Socioeconomic Status and In Utero Fetal Brain Development. JAMA Netw Open 2021; 4:e213526. [PMID: 33779746 PMCID: PMC8008281 DOI: 10.1001/jamanetworkopen.2021.3526] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPORTANCE Children raised in settings with lower parental socioeconomic status are at increased risk for neuropsychological disorders. However, to date, the association between socioeconomic status and fetal brain development remains poorly understood. OBJECTIVE To determine the association between parental socioeconomic status and in vivo fetal brain growth and cerebral cortical development using advanced, 3-dimensional fetal magnetic resonance imaging. DESIGN, SETTING, AND PARTICIPANTS This cohort study of fetal brain development enrolled 144 healthy pregnant women from 2 low-risk community obstetrical hospitals from 2012 through 2019 in the District of Columbia. Included women had a prenatal history without complications that included recommended screening laboratory and ultrasound studies. Exclusion criteria were multiple gestation pregnancy, known or suspected congenital infection, dysmorphic features of the fetus, and documented chromosomal abnormalities. T2-weighted fetal brain magnetic resonance images were acquired. Each pregnant woman was scanned at up to 2 points in the fetal period. Data were analyzed from June through November 2020. EXPOSURES Parental education level and occupation status were documented. MAIN OUTCOMES AND MEASURES Regional fetal brain tissue volume (for cortical gray matter, white matter, cerebellum, deep gray matter, and brainstem) and cerebral cortical features (ie, lobe volume, local gyrification index, and sulcal depth) in the frontal, parietal, temporal, and occipital lobes were calculated. RESULTS Fetal brain magnetic resonance imaging studies were performed among 144 pregnant women (median [interquartile range] age, 32.5 [27.0-36.1] years) with gestational age from 24.0 to 39.4 weeks; 75 fetuses (52.1%) were male, and 69 fetuses (47.9%) were female. Higher parental education level was associated with significantly increased volume in the fetal white matter (mothers: β, 2.86; 95% CI, 1.26 to 4.45; P = .001; fathers: β, 2.39; 95% CI, 0.97 to 3.81; P = .001), deep gray matter (mothers: β, 0.16; 95% CI, 0.002 to 0.32; P = .048; fathers: β, 0.16; 95% CI, 0.02 to 0.31; P = .02), and brainstem (mothers: β, 0.06; 95% CI, 0.02 to 0.10; P = .01; fathers: β, 0.04; 95% CI, 0.004 to 0.08; P = .03). Higher maternal occupation status was associated with significantly increased volume in the fetal white matter (β, 2.07; 95% CI, 0.88 to 3.26; P = .001), cerebellum (β, 0.17; 95% CI, 0.04 to 0.29; P = .01), and brainstem (β, 0.03; 95% CI, 0.001 to 0.07; P = .04), and higher paternal occupation status was associated with significantly increased white matter volume (β, 1.98; 95% CI, 0.71 to 3.25; P < .01). However, higher socioeconomic status was associated with significantly decreased fetal cortical gray matter volume (mothers: β, -0.11; 95% CI, -0.18 to -0.03; P = .01; fathers: β, -0.10; 95% CI, -0.18 to -0.03; P = .01). Higher parental socioeconomic status was associated with increased volumes of 3 brain lobes of white matter: frontal lobe (mothers: β, 0.07; 95% CI, 0.02 to 0.13; P = .01; fathers: β, 0.06; 95% CI, 0.01 to 0.11; P = .03), parietal lobe (mothers: β, 0.07; 95% CI, 0.03 to 0.11; P < .001; fathers: β, 0.06; 95% CI, 0.03 to 0.10; P = .001), and temporal lobe (mothers: β, 0.04; 95% CI, 0.02 to 0.07; P < .001; fathers: β, 0.04; 95% CI, 0.02 to 0.07; P < .001), and maternal SES score was associated with significantly decreased volume in the occipital lobe (β, 0.02; 95% CI, 0.002 to 0.04; P = .03). Higher parental socioeconomic status was associated with decreased cortical local gyrification index (for example, for the frontal lobe, mothers: β, -1.1; 95% CI, -1.9 to -0.3; P = .01; fathers: β, -0.8; 95% CI, -1.6 to -0.1; P = .03) and sulcal depth, except for the frontal lobe (for example, for the parietal lobe, mothers: β, -9.5; 95% CI, -13.8 to -5.3; P < .001; fathers: β, -8.7; 95% CI, -13.0 to -4.4; P < .001). CONCLUSIONS AND RELEVANCE This cohort study found an association between parental socioeconomic status and altered in vivo fetal neurodevelopment. While being born and raised in a lower socioeconomic status setting is associated with poorer neuropsychological, educational, and socioeconomic outcomes in children, these findings suggest that altered prenatal programming may be associated with these outcomes and that future targeted prenatal interventions may be needed.
Collapse
Affiliation(s)
- Yuan-Chiao Lu
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Kushal Kapse
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Nicole Andersen
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Jessica Quistorff
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Catherine Lopez
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Andrea Fry
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Jenhao Cheng
- Department of Quality and Patient Safety, Children's National Hospital, Washington, District of Columbia
| | - Nickie Andescavage
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
- Department of Pediatrics, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia
| | - Yao Wu
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Kristina Espinosa
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Gilbert Vezina
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
| | - Adre du Plessis
- Prenatal Pediatrics Institute, Children's National Hospital, Washington, District of Columbia
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, Washington, District of Columbia
- Department of Pediatrics, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia
| |
Collapse
|
105
|
Xu YH, Wang XX, Wang MJ, Liu YY, Xue Z, Chen JX. Influence of progestational stress on BDNF and NMDARs in the hippocampus of male offspring and amelioration by Chaihu Shugan San. Biomed Pharmacother 2021; 135:111204. [PMID: 33548869 DOI: 10.1016/j.biopha.2020.111204] [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: 08/11/2020] [Revised: 12/13/2020] [Accepted: 12/26/2020] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Progestational stress has been proven to be a risk for the neural development of offspring, especially in the hippocampus. However, whether Chaihu Shugan San (CSS) can ameliorate hippocampal neural development via the regulation of brain-derived neurotrophic factor (BDNF), and N-methyl-D-aspartate receptors (NMDAR) 2A (NR2A) and 2B (NR2B), and the mechanism of such action remains unclear. METHODS Thirty-six female rats were randomly allocated into control, chronic immobilization stress (CIS) and CSS groups according to the random number table, respectively. The male offspring were fed for 21 days after birth then randomly divided into the same three groups (6 rats/group) as the female rats. Female rats, except for the control group, underwent 21-day CIS to established a progestational stress anxiety-like model which was evaluated by body weight, the elevated plus-maze (EPM) test and serum dopamine (DA) measured using an enzyme-linked immunosorbent assay (ELISA). The expression levels of estrogen receptors (ERα/ERβ) and progesterone receptor (PR) in female rat ovaries were quantified by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. The hippocampal tissue in the 21-day offspring was observed by hematoxylin-eosin (HE) staining. The concentration of BDNF, NR2A, and NR2B were measured by RT-qPCR and immunohistochemistry in the CA3 and dentate gyrus (DG) regions of offsprings' hippocampus. RESULTS Compared with the female control group, significant differences in body weight, EPM test and DA concentration were observed in the CIS group, meanwhile, the concentration of ERα (P < 0.05), PR (P < 0.05) and ERβ in the ovaries were decreased. In the offsprings' hippocampus of the CIS group, the chromatin of the nucleus was edge set and with condensed and irregular morphology nucleus, and the cytoplasm was unevenly stained with spaces around the cells, moreover, the expression levels of BDNF, NR2A, and NR2B were also declined (P < 0.05). However, Chaihu Shugan San reversed these changes, especially the BDNF in the DG region (P < 0.05), and NR2A and NR2B in the CA3 and DG region (P < 0.05). CONCLUSIONS CSS could ameliorate the neural development of the hippocampus in offspring damaged by anxiety-like progestational stress in female rats via regulating the expression levels of ERα, ERβ, and PR in female rat ovaries and BDNF, NR2A, and NR2B in the hippocampus of their offspring.
Collapse
MESH Headings
- Animals
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Disease Models, Animal
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Female
- Gestational Age
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Male
- Neurogenesis/drug effects
- Ovary/drug effects
- Ovary/metabolism
- Plant Extracts/pharmacology
- Pregnancy
- Prenatal Exposure Delayed Effects
- Rats, Wistar
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Restraint, Physical
- Signal Transduction
- Stress, Psychological/drug therapy
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
- Rats
Collapse
Affiliation(s)
- Ya-Hui Xu
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Xing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Ming-Jing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Yue-Yun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
106
|
Church JS, Tamayo JM, Ashwood P, Schwartzer JJ. Repeated allergic asthma in early versus late pregnancy differentially impacts offspring brain and behavior development. Brain Behav Immun 2021; 93:66-79. [PMID: 33358979 PMCID: PMC7979463 DOI: 10.1016/j.bbi.2020.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/24/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Stress during pregnancy and maternal inflammation are two common prenatal factors that impact offspring development. Asthma is the leading chronic condition complicating pregnancy and a common source of prenatal stress and inflammation. OBJECTIVE The goal of this study was to characterize the developmental impact of repeated allergic asthma inflammation during pregnancy on offspring behavioral outcomes and brain inflammation. METHODS Pregnant female C57BL/6 mice were sensitized with ovalbumin (OVA) or PBS vehicle control and then randomly assigned to receive daily aerosol exposures to the same OVA or PBS treatment during early, gestational days (GD) 2-GD9, or late pregnancy, GD10-GD17. Maternal sera were collected after the first and last aerosol induction regimen and measured for concentrations of corticosterone, anti-OVA IgE, and cytokine profiles. Juvenile male and female offspring were assessed for locomotor and social behaviors and later as adults assessed for anxiety-like, and marble burying behaviors using a series of behavioral tasks. Offspring brains were evaluated for region-specific differences in cytokine concentrations. RESULTS In early gestation, both PBS and OVA-exposed dams had similar serum corticosterone concentration at the start (GD2) and end (GD9) of daily aerosol inductions. Only OVA-exposed dams showed elevations in cytokines that imply a diverse and robust T helper cell-mediated immune response. Male offspring of early OVA-exposed dams showed decreases in open-arm exploration in the elevated plus maze and increased marble burying without concomitant changes in locomotor activity or social interactions. These behavioral deficits in early OVA-exposed male offspring were associated with lower concentrations of G-CSF, IL-4, IL-7, IFNγ, and TNFα in the hypothalamus. In late gestation, both PBS and OVA-exposed dams had increased corticosterone levels at the end of daily aerosol inductions (GD17) compared to at the start of inductions (GD10). Male offspring from both PBS and OVA-exposed dams in late gestation showed similar decreases in open arm exploration on the elevated plus maze compared to OVA male offspring exposed in early gestation. No behavioral differences were present in female offspring across all treatment groups. However, females of dams exposed to OVA during early gestation displayed similar reductions as males in hypothalamic G-CSF, IL-7, IL-4, and IFNγ. DISCUSSION The inflammatory responses from maternal allergic asthma in early gestation and resulting increases in anxiety-like behavior in males support a link between the timing of prenatal insults and sex-specific developmental outcomes. Moreover, the heightened stress responses in late gestation and concomitant dampened inflammatory response to allergic asthma suggest that interactions between the maternal immune and stress-response systems shape early life fetal programming.
Collapse
Affiliation(s)
- Jamie S Church
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA.
| |
Collapse
|
107
|
Fitzgerald E, Parent C, Kee MZL, Meaney MJ. Maternal Distress and Offspring Neurodevelopment: Challenges and Opportunities for Pre-clinical Research Models. Front Hum Neurosci 2021; 15:635304. [PMID: 33643013 PMCID: PMC7907173 DOI: 10.3389/fnhum.2021.635304] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Pre-natal exposure to acute maternal trauma or chronic maternal distress can confer increased risk for psychiatric disorders in later life. Acute maternal trauma is the result of unforeseen environmental or personal catastrophes, while chronic maternal distress is associated with anxiety or depression. Animal studies investigating the effects of pre-natal stress have largely used brief stress exposures during pregnancy to identify critical periods of fetal vulnerability, a paradigm which holds face validity to acute maternal trauma in humans. While understanding these effects is undoubtably important, the literature suggests maternal stress in humans is typically chronic and persistent from pre-conception through gestation. In this review, we provide evidence to this effect and suggest a realignment of current animal models to recapitulate this chronicity. We also consider candidate mediators, moderators and mechanisms of maternal distress, and suggest a wider breadth of research is needed, along with the incorporation of advanced -omics technologies, in order to understand the neurodevelopmental etiology of psychiatric risk.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Carine Parent
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Michelle Z. L. Kee
- Translational Neuroscience Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Michael J. Meaney
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
- Translational Neuroscience Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
108
|
Wong G, Weir JM, Mishra P, Huynh K, Nijagal B, Gupta V, Broekman BFP, Chong MFF, Chan SY, Tan KH, Tull D, McConville M, Calder PC, Godfrey KM, Chong YS, Gluckman PD, Meaney MJ, Meikle PJ, Karnani N. The placental lipidome of maternal antenatal depression predicts socio-emotional problems in the offspring. Transl Psychiatry 2021; 11:107. [PMID: 33542173 PMCID: PMC7862650 DOI: 10.1038/s41398-021-01208-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 10/06/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
While maternal mental health strongly influences neurodevelopment and health in the offspring, little is known about the determinants of inter-individual variation in the mental health of mothers. Likewise, the in utero biological pathways by which variation in maternal mental health affects offspring development remain to be defined. Previous studies implicate lipids, consistent with a known influence on cognitive and emotional function, but the relevance for maternal mental health and offspring neurodevelopment is unclear. This study characterizes the placental and circulatory lipids in antenatal depression, as well as socio-emotional outcomes in the offspring. Targeted liquid chromatography-mass spectrometry covering 470 lipid species was performed on placenta from 186 women with low (n = 70) or high (n = 116) levels of antenatal depressive symptoms assessed using the Edinburgh Postnatal Depression Scale at 26 weeks' gestation. Child socio-emotional outcomes were assessed from the Child Behavior Check List (CBCL) at 48 months. Seventeen placental lipid species showed an inverse association with antenatal EPDS scores. Specifically, lower levels of phospholipids containing LC-PUFAs: omega-3 docosapentaenoic acid (DPA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and omega-6 arachidonic acid (AA) were significantly associated with depressive symptoms. Additional measurement of LC-PUFA in antenatal plasma samples at mid-gestation confirmed the reduced circulation of these specific fatty acids in mothers. Reduced concentration of the placental phospholipids also predicted poorer socio-emotional outcomes in the offspring. This study provides new insights into the role of the materno-fetal lipid cross-talk as a mechanism linking maternal mental health to that of the offspring. These findings show the potential utility of nutritional approaches among pregnant women with depressive symptoms to reduce offspring risk for later socio-emotional problems.
Collapse
Affiliation(s)
- Gerard Wong
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Jacquelyn M. Weir
- grid.1051.50000 0000 9760 5620Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Priti Mishra
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Kevin Huynh
- grid.1051.50000 0000 9760 5620Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Brunda Nijagal
- grid.1008.90000 0001 2179 088XMetabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Varsha Gupta
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Birit F. P. Broekman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.12380.380000 0004 1754 9227Department of Psychiatry, OLVG and Amsterdam UMC, VU University, Amsterdam, the Netherlands
| | - Mary Foong-Fong Chong
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Saw Swee Hock School of Public Health, National University of Singapore (NUS) and National University Health System (NUHS), Singapore, Singapore
| | - Shiao-Yng Chan
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Kok Hian Tan
- grid.414963.d0000 0000 8958 3388KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Dedreia Tull
- grid.1008.90000 0001 2179 088XMetabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Malcolm McConville
- grid.1008.90000 0001 2179 088XMetabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Philip C. Calder
- grid.5491.90000 0004 1936 9297School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK ,grid.5491.90000 0004 1936 9297NIHR Southampton Biomedical Research Centre, Southampton University Hospital NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Keith M. Godfrey
- grid.5491.90000 0004 1936 9297NIHR Southampton Biomedical Research Centre, Southampton University Hospital NHS Foundation Trust and University of Southampton, Southampton, UK ,grid.5491.90000 0004 1936 9297MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Yap Seng Chong
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Peter D. Gluckman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.9654.e0000 0004 0372 3343Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Michael J. Meaney
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore ,grid.14709.3b0000 0004 1936 8649Sackler Program for Epigenetics & Psychobiology at McGill University, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Peter J. Meikle
- grid.1051.50000 0000 9760 5620Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.
| |
Collapse
|
109
|
The Role of the FOXO1/β 2-AR/p-NF-κB p65 Pathway in the Development of Endometrial Stromal Cells in Pregnant Mice under Restraint Stress. Int J Mol Sci 2021; 22:ijms22031478. [PMID: 33540675 PMCID: PMC7867244 DOI: 10.3390/ijms22031478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/23/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Restraint stress causes various maternal diseases during pregnancy. β2-Adrenergic receptor (β2-AR) and Forkhead transcription factor class O 1 (FOXO1) are critical factors not only in stress, but also in reproduction. However, the role of FOXO1 in restraint stress, causing changes in the β2-AR pathway in pregnant mice, has been unclear. The aim of this research was to investigate the β2-AR pathway of restraint stress and its impact on the oxidative stress of the maternal uterus. In the study, maternal mice were treated with restraint stress by being restrained in a transparent and ventilated device before sacrifice on Pregnancy Day 5 (P5), Pregnancy Day 10 (P10), Pregnancy Day 15 (P15), and Pregnancy Day 20 (P20) as well as on Non-Pregnancy Day 5 (NP5). Restraint stress augmented blood corticosterone (CORT), norepinephrine (NE), and blood glucose levels, while oestradiol (E2) levels decreased. Moreover, restraint stress increased the mRNA levels of the FOXO family, β2-AR, and even the protein levels of FOXO1 and β2-AR in the uterus and ovaries. Furthermore, restraint stress increased uterine oxidative stress level. In vitro, the protein levels of FOXO1 were also obviously increased when β2-AR was activated in endometrial stromal cells (ESCs). In addition, phosphorylated-nuclear factor kappa-B p65 (p-NF-κB p65) and its target genes decreased significantly when FOXO1 was inhibited. Overall, it can be said that the β2-AR/FOXO1/p-NF-κB p65 pathway was activated when pregnant mice were under restraint stress. This study provides a scientific basis for the origin of psychological stress in pregnant women.
Collapse
|
110
|
Lopizzo N, Mazzelli M, Zonca V, Begni V, D'Aprile I, Cattane N, Pariante CM, Riva MA, Cattaneo A. Alterations in 'inflammatory' pathways in the rat prefrontal cortex as early biological predictors of the long-term negative consequences of exposure to stress early in life. Psychoneuroendocrinology 2021; 124:104794. [PMID: 33429258 DOI: 10.1016/j.psyneuen.2020.104794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022]
Abstract
Early life stress, especially when experienced during the first period of life, affects the brain developmental trajectories leading to an enhanced vulnerability for stress-related psychiatric disorders later in life. Although both clinical and preclinical studies clearly support this association, the biological pathways deregulated by such exposure, and the effects in shaping the neurodevelopmental trajectories, have so far been poorly investigated. By using the prenatal stress (PNS) model, a well-established rat model of early life stress, we performed transcriptomic analyses in the prefrontal cortex of rats exposed or not to PNS and sacrificed at different postnatal days (PNDs 21, 40, 62). We first investigated the long-lasting mechanisms and pathways affected in the PFC. We have decided to focus on the prefrontal cortex because we have previously shown that this brain region is highly sensitive to PNS exposure. We found that adult animals exposed to PNS show alterations in 389 genes, mainly involved in stress and inflammatory signalling. We then wanted to establish whether PNS exposure could also affect the neurodevelopmental trajectories in order to identify the most critical temporal window. We found that PNS rats show the most significant changes during adolescence (between PND 40 versus PND 21), with alterations of several pathways related to stress, inflammation and metabolism, which were maintained until adulthood.
Collapse
Affiliation(s)
- Nicola Lopizzo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Monica Mazzelli
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Valentina Zonca
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy.
| |
Collapse
|
111
|
Guan SZ, Fu YJ, Zhao F, Liu HY, Chen XH, Qi FQ, Liu ZH, Ng TB. The mechanism of enriched environment repairing the learning and memory impairment in offspring of prenatal stress by regulating the expression of activity-regulated cytoskeletal-associated and insulin-like growth factor-2 in hippocampus. Environ Health Prev Med 2021; 26:8. [PMID: 33451279 PMCID: PMC7811238 DOI: 10.1186/s12199-020-00929-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/27/2020] [Indexed: 01/03/2023] Open
Abstract
Background Prenatal stress can cause neurobiological and behavioral defects in offspring; environmental factors play a crucial role in regulating the development of brain and behavioral; this study was designed to test and verify whether an enriched environment can repair learning and memory impairment in offspring rats induced by prenatal stress and to explore its mechanism involving the expression of insulin-like growth factor-2 (IGF-2) and activity-regulated cytoskeletal-associated protein (Arc) in the hippocampus of the offspring. Methods Rats were selected to establish a chronic unpredictable mild stress (CUMS) model during pregnancy. Offspring were weaned on 21st day and housed under either standard or an enriched environment. The learning and memory ability were tested using Morris water maze and Y-maze. The expression of IGF-2 and Arc mRNA and protein were respectively measured by using RT-PCR and Western blotting. Results There was an elevation in the plasma corticosterone level of rat model of maternal chronic stress during pregnancy. Maternal stress’s offspring exposed to an enriched environment could decrease their plasma corticosterone level and improve their weight. The offspring of maternal stress during pregnancy exhibited abnormalities in Morris water maze and Y-maze, which were improved in an enriched environment. The expression of IGF-2, Arc mRNA, and protein in offspring of maternal stress during pregnancy was boosted and some relationships existed between these parameters after being exposed enriched environment. Conclusions The learning and memory impairment in offspring of prenatal stress can be rectified by the enriched environment, the mechanism of which is related to the decreasing plasma corticosterone and increasing hippocampal IGF-2 and Arc of offspring rats following maternal chronic stress during pregnancy.
Collapse
Affiliation(s)
- Su-Zhen Guan
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Department of Occupational Health and Environmental Health, College of Public Health, Xinjiang Medical University, Urumqi, 830011, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - You-Juan Fu
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Feng Zhao
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Hong-Ya Liu
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Xiao-Hui Chen
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Fa-Qiu Qi
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Zhi-Hong Liu
- School of Public Health and Management, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China. .,Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
112
|
Broeks CW, Choenni V, Kok R, van der Voorn B, de Kruijff I, van den Akker ELT, van Rossum EFC, Hoogendijk WJG, Hillegers MHJ, Kamperman AM, Lambregtse-Van den Berg MP. An exploratory study of perinatal hair cortisol concentrations in mother-infant dyads with severe psychiatric disorders versus healthy controls. BJPsych Open 2021; 7:e28. [PMID: 33407971 PMCID: PMC8058941 DOI: 10.1192/bjo.2020.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Maternal psychopathology during pregnancy is associated with negative outcomes in offspring. Increased placental transfer of maternal cortisol may contribute to mediate this association. Hair cortisol concentrations (HCCs) appear to be a good biomarker of long-term prenatal stress exposure. Little is known about the associations between severe maternal psychopathology and perinatal infant HCCs. AIMS We assessed HCCs in the perinatal period in mother-infant dyads with and without severe psychiatric disorders. METHOD We examined group differences in HCCs of mother-infant dyads (n = 18) subjected to severe maternal psychiatric disorders versus healthy control dyads (n = 27). We assessed the correlation of HCCs between mother and infant within both groups, and the association between current maternal symptoms and HCCs in patient dyads. RESULTS Median (interquartile range) and distribution of HCC differed in patients compared with control mothers (U = 468.5, P = 0.03). HCCs in infants of patients did not differ from control infants (U = 250.0, P = 0.67). Subsequently, we found that HCCs within healthy control dyads were correlated (n = 27, r 0.55 (0.14), P = 0.003), but were not within patient dyads (n = 18, r 0.082 (0.13), P = 0.746). HCCs in infants of patients showed a positive correlation with maternal symptoms (n = 16, r = 0.63 (0.06), P = 0.008). CONCLUSIONS These preliminary findings suggest that infant HCC reflect perinatal stress exposure. In infants, these early differences could influence lifetime hypothalamic-pituitary-adrenal axis functioning, which might be associated with increased susceptibility to later disease.
Collapse
Affiliation(s)
- Carlinde W Broeks
- Department of Psychiatry, Erasmus University Medical Center, the Netherlands; and Department of Psychiatry, Arkin Institute for Mental Health, the Netherlands
| | - Vandhana Choenni
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands
| | - Rianne Kok
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, the Netherlands
| | - Bibian van der Voorn
- Department of Pediatric Endocrinology, Obesity Center CGG, Erasmus MC-Sophia Children's Hospital, the Netherlands; and Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, the Netherlands
| | - Ineke de Kruijff
- Department of Pediatrics, St Antonius Hospital Nieuwegein, the Netherlands
| | | | - Elisabeth F C van Rossum
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, the Netherlands
| | | | - Manon H J Hillegers
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands
| | - Astrid M Kamperman
- Department of Psychiatry, Erasmus University Medical Center, the Netherlands; and Epidemiological and Social Psychiatric Research Institute, Erasmus University Medical Center, the Netherlands
| | - Mijke P Lambregtse-Van den Berg
- Department of Psychiatry, Erasmus University Medical Center, the Netherlands; and Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands
| |
Collapse
|
113
|
Bush NR, Savitz J, Coccia M, Jones-Mason K, Adler N, Boyce WT, Laraia B, Epel E. Maternal Stress During Pregnancy Predicts Infant Infectious and Noninfectious Illness. J Pediatr 2021; 228:117-125.e2. [PMID: 32827529 PMCID: PMC7752845 DOI: 10.1016/j.jpeds.2020.08.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/31/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To examine the association between prenatal stress and infant physical health in the first year of life within an understudied, racially and ethnically diverse, highly stressed community sample. We expected that greater stress exposure would predict higher rates of infant illness. STUDY DESIGN Low-income, racially/ethnically diverse, overweight women with low medical risk pregnancies were recruited (2011-2014) during pregnancy. Pregnancy Stressful Life Events were assessed retrospectively (mean, 11.88 months postpartum). Perceived stress was assessed twice during pregnancy (at a mean of 17.4 weeks and again at a mean of 25.6 weeks) and at 6 months postpartum. Women with live births (n = 202) were invited; 162 consented to the offspring study. Medical records from pediatric clinics and emergency departments for 148 infants were abstracted for counts of total infectious illnesses, total noninfectious illness, and diversity of illnesses over the first year of life. RESULTS The final analytic sample included 109 women (mean age, 28.08 years) and their infants. In covariate-adjusted negative binomial models, maternal perceptions of stress across pregnancy were positively associated with infant illness. Each 1-point increase in average stress was associated with a 38% increase in incidence of infant infections (Incidence rate ratio, 1.38; 95% CI, 1.01-1.88; P < .05), a 73% increase in noninfectious illness (IRR, 1.73; 95% CI, 1.34-2.23; P < .05), and a 53% increase in illness diversity (IRR, 1.53; 95% CI, 1.25, 1.88; P < .01); effect sizes were larger for perceived stress later in pregnancy. Stressful life events count and postnatal stress were not uniquely associated with illness. CONCLUSIONS In line with recommendations from the American Academy of Pediatrics to screen for maternal perinatal depression, screening and support for stress reduction during pregnancy may benefit both maternal and child health.
Collapse
Affiliation(s)
- Nicole R. Bush
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry,Department of Pediatrics, Division of Developmental Medicine, University of California, San Francisco
| | - Jennifer Savitz
- Departments of Medicine and Clinical Pharmacy, University of California, San Francisco, CA,University of Washington and Seattle Children's Hospital, Seattle, WA
| | - Michael Coccia
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry
| | - Karen Jones-Mason
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry
| | - Nancy Adler
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry
| | - W. Thomas Boyce
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry,Department of Pediatrics, Division of Developmental Medicine, University of California, San Francisco
| | - Barbara Laraia
- School of Public Health, University of California, Berkeley, CA
| | - Elissa Epel
- Weill Institute for Neurosciences, Center for Health and Community, Department of Psychiatry
| |
Collapse
|
114
|
Wu Y, Zhang H, Wang C, Broekman BFP, Chong YS, Shek LP, Gluckman PD, Meaney MJ, Fortier MV, Qiu A. Inflammatory modulation of the associations between prenatal maternal depression and neonatal brain. Neuropsychopharmacology 2021; 46:470-477. [PMID: 32688365 PMCID: PMC7852623 DOI: 10.1038/s41386-020-0774-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/26/2020] [Accepted: 07/13/2020] [Indexed: 11/09/2022]
Abstract
Inflammatory signaling has a role in sensing intrauterine environment, which may be moderators in altering fetal brain development upon maternal environment. This study integrated cytokine transcriptome of post-mortem fetal brains, neonatal brain imaging and genetic variants (n = 161) to examine whether cytokines are candidates for modulating the relationship between prenatal maternal depression and fetal brain development. This study obtained the transcriptome data of 208 cytokine genes in 12 fetal brain regions from the BrainSpan database. We also included 161 mother-child dyads with prenatal maternal depressive symptoms assessed at 26 weeks of gestation, cytokine genotype data extracted from umbilical cord specimens, and neonatal brain images from a longitudinal prospective birth cohort. We revealed that 22 cytokine genes are expressed in specific brain regions in utero, whose variants have roles in modulating the effects of the prenatal environment on the accelerated fetal development of the hippocampus, auditory, parietal, orbitofrontal, and dorsal prefrontal cortex. Neonates high in the genetic expression score (GES) of TNFRSF19 and IL17RB showed a larger right hippocampal volume, high in the GES of BMPR1B showed the thicker thickness of the sensorimotor cortex, and high in the GES of IL1RAP and CXCR4 demonstrated the thicker thickness of the dorsal and orbital prefrontal cortex in relation with greater prenatal maternal depressive symptoms. Our findings suggest that in humans, the cytokine genes are expressed in a brain region-specific manner in utero and may have potential roles in modulating the fetal development of the corresponding brain regions in response to the maternal environment.
Collapse
Affiliation(s)
- Yonghui Wu
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Han Zhang
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Changqing Wang
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Birit F. P. Broekman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Yap-Seng Chong
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Lynette P. Shek
- grid.4280.e0000 0001 2180 6431Department of Pediatrics, Khoo Teck Puat – National University Children’s Medical Institute, National University of Singapore, Singapore, Singapore
| | - Peter D. Gluckman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Michael J. Meaney
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics and Mental Health, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada
| | - Marielle V. Fortier
- grid.414963.d0000 0000 8958 3388Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore. .,The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
115
|
Kimura H, Mori D, Aleksic B, Ozaki N. Elucidation of molecular pathogenesis and drug development for psychiatric disorders from rare disease-susceptibility variants. Neurosci Res 2020; 170:24-31. [PMID: 33316300 DOI: 10.1016/j.neures.2020.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 10/22/2022]
Abstract
Recent rapid progress in genome analysis and large-scale consortia has made it possible to discover variants with a variety of allele frequencies and effect sizes associated with psychiatric disorders. Among psychiatric disorder-susceptibility variants, rare variants with large effect sizes detected by sequencing analysis or array comparative genomic hybridization would be particularly useful for elucidating pathophysiology by developing disease models, such as genome-edited mouse or induced pluripotent stem cells. In the last decade, investigations of rare variants with large effect size have revealed an important role of neurodevelopment in the pathogenesis of psychiatric disorders. In future research, integration of recent evidence concerning the contribution of the immune system or gut microbiota will enhance our understanding of psychiatric disorders and facilitate novel drug development.
Collapse
Affiliation(s)
- Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain & Mind Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain & Mind Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
116
|
Yang F, Zhang F, Ji X, Jiang X, Xue M, Yu H, Hu X, Bao Z. Secretory galectin-3 induced by glucocorticoid stress triggers stemness exhaustion of hepatic progenitor cells. J Biol Chem 2020; 295:16852-16862. [PMID: 32989051 PMCID: PMC7864077 DOI: 10.1074/jbc.ra120.012974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 09/17/2020] [Indexed: 12/18/2022] Open
Abstract
Adult progenitor cell populations typically exist in a quiescent state within a controlled niche environment. However, various stresses or forms of damage can disrupt this state, which often leads to dysfunction and aging. We built a glucocorticoid (GC)-induced liver damage model of mice, found that GC stress induced liver damage, leading to consequences for progenitor cells expansion. However, the mechanisms by which niche factors cause progenitor cells proliferation are largely unknown. We demonstrate that, within the liver progenitor cells niche, Galectin-3 (Gal-3) is responsible for driving a subset of progenitor cells to break quiescence. We show that GC stress causes aging of the niche, which induces the up-regulation of Gal-3. The increased Gal-3 population increasingly interacts with the progenitor cell marker CD133, which triggers focal adhesion kinase (FAK)/AMP-activated kinase (AMPK) signaling. This results in the loss of quiescence and leads to the eventual stemness exhaustion of progenitor cells. Conversely, blocking Gal-3 with the inhibitor TD139 prevents the loss of stemness and improves liver function. These experiments identify a stress-dependent change in progenitor cell niche that directly influence liver progenitor cell quiescence and function.
Collapse
Affiliation(s)
- Fan Yang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fan Zhang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Xueying Ji
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Xin Jiang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Mengjuan Xue
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Huiyuan Yu
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Xiaona Hu
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Zhijun Bao
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
117
|
Antonson AM, Evans MV, Galley JD, Chen HJ, Rajasekera TA, Lammers SM, Hale VL, Bailey MT, Gur TL. Unique maternal immune and functional microbial profiles during prenatal stress. Sci Rep 2020; 10:20288. [PMID: 33219314 PMCID: PMC7679384 DOI: 10.1038/s41598-020-77265-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Maternal stress during pregnancy is widespread and is associated with poor offspring outcomes, including long-term mental health issues. Prenatal stress-induced fetal neuroinflammation is thought to underlie aberrant neurodevelopment and to derive from a disruption in intrauterine immune homeostasis, though the exact origins are incompletely defined. We aimed to identify divergent immune and microbial metagenome profiles of stressed gestating mice that may trigger detrimental inflammatory signaling at the maternal-fetal interface. In response to stress, maternal glucocorticoid circuit activation corresponded with indicators of systemic immunosuppression. At the maternal-fetal interface, density of placental mononuclear leukocytes decreased with stress, yet maternal whole blood leukocyte analysis indicated monocytosis and classical M1 phenotypic shifts. Genome-resolved microbial metagenomic analyses revealed reductions in genes, microbial strains, and metabolic pathways in stressed dams that are primarily associated with pro-inflammatory function. In particular, disrupted Parasutterella excrementihominis appears to be integral to inflammatory and metabolic dysregulation during prenatal stress. Overall, these perturbations in maternal immunological and microbial regulation during pregnancy may displace immune equilibrium at the maternal-fetal interface. Notably, the absence of and reduction in overt maternal inflammation during stress indicates that the signaling patterns driving fetal outcomes in this context are more nuanced and complex than originally anticipated.
Collapse
Affiliation(s)
- Adrienne M Antonson
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Morgan V Evans
- Environmental Health Sciences Division, College of Public Health, The Ohio State University, Columbus, OH, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Jeffrey D Galley
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Helen J Chen
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Medical Scientist Training Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Therese A Rajasekera
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Environmental Health Sciences Division, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Sydney M Lammers
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Barnes Medical Student Research Scholarship Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Vanessa L Hale
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Michael T Bailey
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Center for Microbial Pathogenesis, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tamar L Gur
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Medical Scientist Training Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, 120A Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, USA.
| |
Collapse
|
118
|
Activated microglia cause metabolic disruptions in developmental cortical interneurons that persist in interneurons from individuals with schizophrenia. Nat Neurosci 2020; 23:1352-1364. [PMID: 33097921 PMCID: PMC7769122 DOI: 10.1038/s41593-020-00724-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
The mechanisms by which prenatal immune activation increase risk for neuropsychiatric disorders are unclear. Here, we generated developmental cortical interneurons (cINs), known to be affected in schizophrenia (SCZ) when matured, from induced pluripotent stem cells (iPSCs) from healthy controls (HC) and SCZ patients, and cocultured them with or without activated microglia. Coculture with activated microglia disturbed metabolic pathways, as indicated by unbiased transcriptome analysis, and impaired mitochondrial function, arborization, synapse formation and synaptic GABA release. Deficits in mitochondrial function and arborization were reversed by Alpha Lipoic Acid/Acetyl-L-Carnitine (ALA/ALC) treatments that boost mitochondrial function. Notably, activated microglia-conditioned medium altered metabolism in cINs and HC-derived iPSCs but not in SCZ-patient-derived iPSCs or in glutamatergic neurons. After removal of activated microglia-conditioned medium, SCZ cINs but not HC cINs showed prolonged metabolic deficits, suggesting an interaction between SCZ genetic backgrounds and environmental risk factors.
Collapse
|
119
|
Environmental regulation of the chloride transporter KCC2: switching inflammation off to switch the GABA on? Transl Psychiatry 2020; 10:349. [PMID: 33060559 PMCID: PMC7562743 DOI: 10.1038/s41398-020-01027-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Chloride homeostasis, the main determinant factor for the dynamic tuning of GABAergic inhibition during development, has emerged as a key element altered in a wide variety of brain disorders. Accordingly, developmental disorders such as schizophrenia, Autism Spectrum Disorder, Down syndrome, epilepsy, and tuberous sclerosis complex (TSC) have been associated with alterations in the expression of genes codifying for either of the two cotransporters involved in the excitatory-to-inhibitory GABA switch, KCC2 and NKCC1. These alterations can result from environmental insults, including prenatal stress and maternal separation which share, as common molecular denominator, the elevation of pro-inflammatory cytokines. In this review we report and systemize recent research articles indicating that different perinatal environmental perturbations affect the expression of chloride transporters, delaying the developmental switch of GABA signaling, and that inflammatory cytokines, in particular interleukin 1β, may represent a key causal factor for this phenomenon. Based on literature data, we provide therefore a unifying conceptual framework, linking environmental hits with the excitatory-to-inhibitory GABA switch in the context of brain developmental disorders.
Collapse
|
120
|
Prenatal pregnancy-related anxiety predicts boys' ADHD symptoms via placental C-reactive protein. Psychoneuroendocrinology 2020; 120:104797. [PMID: 32682173 DOI: 10.1016/j.psyneuen.2020.104797] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/15/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022]
Abstract
Many modes of stress (i.e. life events, catastrophic events) during pregnancy have been found to increase the risk of externalizing behaviors, and probably in a sex-specific way. Maternal immune activation may be the sex-difference mechanism, but direct evidence that assess three factors in conjunction -- maternal stress, maternal immune activation, and offspring neurodevelopment --from human beings is lacking. This prospective study followed 2926 pregnant women from early pregnancy to 36 months after delivery. Pregnancy-related anxiety symptoms assessment was completed three times using the Pregnancy-Related Anxiety Questionnaire; child attention deficit hyperactivity disorder (ADHD) symptoms were assessed by the parent version of the Conners' Hyperactivity Index. More importantly, nine inflammatory cytokines were detected in placental tissues for the sex-difference mechanism investigation. Our results showed that after controlling for confounding factors, pregnancy-related anxiety during at least two trimesters of pregnancy increased the risk of ADHD for boys (adjusted odds ratio (aOR) = 3.37, 95 % confidence interval (95 % CI) = 1.78-6.38), but not for girls (aOR = 1.02, 95 %CI = 0.44-2.38), which confirmed previous findings. Besides, the structural equation models revealed that placental C-reactive protein (CRP) mRNA expression significantly mediated the association between pregnancy-related anxiety and ADHD for boys (indirect effect: β = 0.025, P = 0.022), but not for girls (indirect effect: β = 0.005, P = 0.589). This prospective study suggested that frequent pregnancy-related anxiety during pregnancy and its induced-placental inflammation partially contributed to the sex-bias of ADHD symptoms.
Collapse
|
121
|
Abdoli A, Falahi S, Kenarkoohi A, Shams M, Mir H, Jahromi MAM. The COVID-19 pandemic, psychological stress during pregnancy, and risk of neurodevelopmental disorders in offspring: a neglected consequence. J Psychosom Obstet Gynaecol 2020; 41:247-248. [PMID: 32380881 DOI: 10.1080/0167482x.2020.1761321] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Amir Abdoli
- Department of Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.,Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Azra Kenarkoohi
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Morteza Shams
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamed Mir
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mirza Ali Mofazzal Jahromi
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.,Department of Advanced Medical Sciences and Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.,Department of Laboratory Sciences, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
122
|
Patel S, Dale RC, Rose D, Heath B, Nordahl CW, Rogers S, Guastella AJ, Ashwood P. Maternal immune conditions are increased in males with autism spectrum disorders and are associated with behavioural and emotional but not cognitive co-morbidity. Transl Psychiatry 2020; 10:286. [PMID: 32796821 PMCID: PMC7429839 DOI: 10.1038/s41398-020-00976-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Epidemiological and animal research shows that maternal immune activation increases the risk of autism spectrum disorders (ASD) in offspring. Emerging evidence suggests that maternal immune conditions may play a role in the phenotypic expression of neurodevelopmental difficulties in children with ASD and this may be moderated by offspring sex. This study aimed to investigate whether maternal immune conditions were associated with increased severity of adverse neurodevelopmental outcomes in children with ASD. Maternal immune conditions were examined as predictors of ASD severity, behavioural and emotional well-being, and cognitive functioning in a cohort of 363 children with ASD (n = 363; 252 males, 111 females; median age 3.07 [interquartile range 2.64-3.36 years]). We also explored whether these outcomes varied between male and female children. Results showed that maternal asthma was the most common immune condition reported in mothers of children with ASD. A history of maternal immune conditions (p = 0.009) was more common in male children with ASD, compared to female children. Maternal immune conditions were associated with increased behavioural and emotional problems in male and female children. By contrast, maternal immune conditions were not associated with decreased cognitive function. The findings demonstrate that MIA may influence the expression of symptoms in children with ASD and outcomes may vary between males and females.
Collapse
Affiliation(s)
- Shrujna Patel
- grid.1013.30000 0004 1936 834XAutism Clinic for Translational Research, Brain and Mind Centre, Children’s Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW Australia
| | - Russell C. Dale
- grid.1013.30000 0004 1936 834XKids Neuroscience Centre, The Children’s Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Westmead, NSW Australia
| | - Destanie Rose
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology and MIND Institute, UC Davis, Davis, CA USA
| | - Brianna Heath
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Christine W. Nordahl
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Sally Rogers
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Adam J. Guastella
- grid.1013.30000 0004 1936 834XAutism Clinic for Translational Research, Brain and Mind Centre, Children’s Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW Australia
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology and MIND Institute, UC Davis, Davis, CA, USA.
| |
Collapse
|
123
|
Meyer JS, Novak MA. Assessment of prenatal stress-related cortisol exposure: focus on cortisol accumulation in hair and nails. Dev Psychobiol 2020; 63:409-436. [PMID: 32783213 DOI: 10.1002/dev.22021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/02/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
Prenatal stress adversely affects offspring development. Although cortisol is hypothesized to be a key mediator of stress-induced developmental deficits, determining the amount of fetal cortisol exposure produced by maternal stress has proved challenging. Current approaches, such as measuring cortisol concentrations in maternal plasma, saliva, or urine, amniotic fluid, fetal plasma, or cord blood, all have significant limitations for assessing cumulative fetal cortisol exposure over time. A recently emerging approach is to measure cortisol concentrations in maternal hair and/or newborn hair or nail samples. Maternal hair cortisol potentially shows long-term production across each trimester of pregnancy, whereas neonatal hair or nail cortisol is thought to reflect mainly third trimester hormone accumulation. This review first describes fetal adrenocortical development, placental cortisol metabolism, and the various sources of fetal cortisol exposure across pregnancy. We then summarize the results obtained from "classical" methods of assessing prenatal cortisol exposure prior to the advent of hair and nail cortisol measurement. Lastly, we discuss the initial development and validation of the hair cortisol methodology, its subsequent application to studies of chronic stress, and recent findings regarding maternal and neonatal hair or nail cortisol concentrations in relation to prenatal stress and other variables of interest.
Collapse
Affiliation(s)
- Jerrold S Meyer
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Program, University of Massachusetts, Amherst, MA, USA
| | - Melinda A Novak
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Program, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
124
|
McLean MA, Simcock G, Elgbeili G, Laplante DP, Kildea S, Hurrion E, Lequertier B, Cobham VE, King S. Disaster-related prenatal maternal stress, and childhood HPA-axis regulation and anxiety: The QF2011 Queensland Flood Study. Psychoneuroendocrinology 2020; 118:104716. [PMID: 32479967 DOI: 10.1016/j.psyneuen.2020.104716] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The fetal programming hypothesis suggests that prenatal maternal stress (PNMS) influences aspects of fetal development, such as the Hypothalamic Pituitary Adrenal (HPA) axis, enhancing susceptibility to emotional problems. No study (to our knowledge) has investigated this pathway considering development of preschool anxiety symptoms. Using data from the Queensland Flood study (QF2011), our objective was to determine whether toddler HPA-axis functioning mediated the association between aspects of flood-related PNMS and child anxiety symptoms at 4-years, and whether relationships were moderated by the timing of the stressor in utero or by the child's sex. METHODS Women, pregnant during the 2011 Queensland floods (N = 230), were recruited soon afterwards and completed questionnaires regarding their objective hardship (e.g., loss of personal property), subjective distress (post-traumatic-like symptoms) and cognitive appraisal of the disaster. At 16 months, indexes of the child's diurnal cortisol rhythm (awakening response, total daily output, diurnal slope [N = 80]), and stress reactivity (N = 111), were obtained. At 4-years, N = 117 mothers reported on their own mood and their children's anxiety symptoms; of these, N = 80 also had valid child cortisol reactivity data, and N = 64 had diurnal cortisol rhythm data. RESULTS A greater cortisol awakening response at 16 months mediated the relationship between subjective PNMS and anxiety symptoms at 4-years. Greater toddler daily cortisol secretion predicted more anxiety symptoms, independent of PNMS. The laboratory stressor did not elicit a cortisol response. PNMS effects were not dependent upon child sex nor on gestational timing of flood exposure. CONCLUSIONS Indexes of diurnal cortisol in toddlerhood may represent vulnerability for anxiety symptoms in preschoolers, both independent of, and following, exposure to disaster-related prenatal maternal subjective distress.
Collapse
Affiliation(s)
- Mia A McLean
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia; School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Gabrielle Simcock
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia; School of Psychology, The University of Queensland, Brisbane, Queensland, Australia; Thompson Institute, University of Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Guillaume Elgbeili
- Schizophrenia and Neurodevelopmental Disorders Research, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - David P Laplante
- Schizophrenia and Neurodevelopmental Disorders Research, Douglas Mental Health University Institute, Verdun, Quebec, Canada; Centre for Child Development and Mental Health, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Sue Kildea
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia; Molly Wardaguga Research Center, School of Nursing and Midwifery, Charles Darwin University, Brisbane, Queensland, Australia
| | - Elizabeth Hurrion
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia
| | - Belinda Lequertier
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia; School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Vanessa E Cobham
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia; School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Suzanne King
- Thompson Institute, University of Sunshine Coast, Sippy Downs, Queensland, Australia; Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
125
|
Daaboul J, Tamouza R, Leboyer M. [Immunopsychiatry and SARS-CoV-2 pandemic: Links and possible consequences]. Encephale 2020; 47:151-156. [PMID: 32928535 PMCID: PMC7373027 DOI: 10.1016/j.encep.2020.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The SARS-CoV-2 (or COVID-19) pandemic has been propagating since December 2019, inducing a drastic increase in the prevalence of anxious and depressive disorders in the general population. Psychological trauma can partly explain these disorders. However, since psychiatric disorders also have an immuno-inflammatory component, the direct effects of the virus on the host's immune system, with a marked inflammatory response, but also the secondary inflammation to these psychosocial stressors, may cause the apparition or the worsening of psychiatric disorders. We describe here the probable immunopsychiatric consequences of the SARS-CoV-2 pandemic, to delineate possible screening actions and care that could be planned. METHOD Data from previous pandemics, and existing data on the psychopathological consequences of the SARS-CoV-2 pandemic, allowed us to review the possible immunopsychiatric consequences of the SARS-CoV-2 pandemic, on the gestational environment, with the risk of consecutive neurodevelopmental disorders for the fetus on one hand, on the children and adults directly infected being at increased risks of psychiatric disorders on the other hand. RESULTS As in previous pandemics, the activation of the immune system due to psychological stress and/or to infection during pregnancy, might lead to an increased risk of neurodevelopmental disorders for the fetus (schizophrenia and autism spectrum disorders). Furthermore, in individuals exposed to psychological trauma and/or infected by the virus, the risk of psychiatric disorders, especially mood disorders, is probably increased. CONCLUSION In this context, preventive measures and specialized care are necessary. Thus, it is important to propose a close follow-up to the individuals who have been infected by the virus, in order to set up the earliest care possible. Likewise, in pregnant women, screening of mood disorders during the pregnancy or the postpartum period must be facilitated. The follow-up of the babies born during the pandemic must be strengthened to screen and care for possible neurodevelopmental disorders.
Collapse
Affiliation(s)
- J Daaboul
- Université de Lille, CHU de Lille, Lille, France; DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France
| | - R Tamouza
- DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France; Fondation FondaMental, Créteil, France; Université Paris Est Créteil, UPEC, Inserm, U955, équipe 15 neuro-psychiatrie translationnelle, Institut Mondor de Recherche Biomédicale, IMRB, Créteil, France
| | - M Leboyer
- DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France; Fondation FondaMental, Créteil, France; Université Paris Est Créteil, UPEC, Inserm, U955, équipe 15 neuro-psychiatrie translationnelle, Institut Mondor de Recherche Biomédicale, IMRB, Créteil, France.
| |
Collapse
|
126
|
Garcia-Rizo C, Bitanihirwe BKY. Implications of early life stress on fetal metabolic programming of schizophrenia: A focus on epiphenomena underlying morbidity and early mortality. Prog Neuropsychopharmacol Biol Psychiatry 2020; 101:109910. [PMID: 32142745 DOI: 10.1016/j.pnpbp.2020.109910] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/16/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022]
Abstract
The fetal origin of adult disease hypothesis postulates that a stressful in utero environment can have deleterious consequences on fetal programming, potentially leading to chronic disease in later life. Factors known to impact fetal programming include the timing, intensity, duration and nature of the external stressor during pregnancy. As such, dynamic modulation of fetal programming is heavily involved in shaping health throughout the life course, possibly by influencing metabolic parameters including insulin action, hypothalamic-pituitary-adrenal activity and immune function. The ability of prenatal insults to program adult disease is likely to occur as a result of reduced functional capacity in key organs-a "thrifty" phenotype-where more resources are re-allocated to preserve critical organs such as the brain. Notably, it has been postulated that the manifestation of neuropsychiatric disorders in individuals priorly exposed to prenatal stress may arise from the interaction between hereditary factors and the intrauterine environment, which together precipitate disease onset by disrupting the trajectory of normal brain development. In this review we discuss the evidence linking prenatal programming to neuropsychiatric disorders, mainly schizophrenia, via a "Thrifty psychiatric phenotype" concept. We start by outlining the conception of the thrifty psychiatric phenotype. Next, we discuss the convergence of potential mechanistic pathways through which prenatal insults may trigger epigenetic changes that contribute to the increased morbidity and early mortality observed in neuropsychiatric disorders. Finally, we touch on the public health importance of fetal programming for these disorders. We conclude by providing a brief outlook on the future of this evolving field of research.
Collapse
Affiliation(s)
- Clemente Garcia-Rizo
- Barcelona Clinic Schizophrenia Unit, Neuroscience Institute, Hospital Clinic, Barcelona, Spain; Institute of Biomedical Research Agusti Pi iSunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| | - Byron K Y Bitanihirwe
- Centre for Global Health, Trinity College Dublin, Dublin, Ireland; Department of Psychology, Trinity College Dublin, Dublin, Ireland; School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
127
|
Li R, Yin T, Fang F, Li Q, Chen J, Wang Y, Hao Y, Wu G, Duan P, Wang Y, Cheng D, Zhou Q, Zafar MI, Xiong C, Li H, Yang J, Qiao J. Potential risks of SARS-CoV-2 infection on reproductive health. Reprod Biomed Online 2020; 41:89-95. [PMID: 32466994 PMCID: PMC7192111 DOI: 10.1016/j.rbmo.2020.04.018] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022]
Abstract
The outbreak of 2019 novel coronavirus disease (COVID-19) has become a major pandemic threat worldwide. Such a public health emergency can greatly impact various aspects of people's health and lives. This paper focuses on its potential risks for reproductive health, including the reproductive system and its functioning, as well as gamete and embryo development, which could be affected by the virus itself, drug treatments, chemical disinfectants and psychological effects related to panic during the COVID-19 outbreak.
Collapse
Affiliation(s)
- Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology Beijing, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China
| | - Fang Fang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Hubei, China
| | - Qin Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology Beijing, China
| | - Jiao Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China
| | - Yixin Wang
- Department of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health MA, USA
| | - Yongxiu Hao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology Beijing, China
| | - Gengxiang Wu
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China
| | - Peng Duan
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine Hubei, China
| | - Yuanyuan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology Beijing, China
| | - Dan Cheng
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China
| | - Qi Zhou
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China
| | - Mohammad Ishraq Zafar
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology Hubei, China
| | | | - Honggang Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology Hubei, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University Hubei, China.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology Beijing, China.
| |
Collapse
|
128
|
Chen HJ, Antonson AM, Rajasekera TA, Patterson JM, Bailey MT, Gur TL. Prenatal stress causes intrauterine inflammation and serotonergic dysfunction, and long-term behavioral deficits through microbe- and CCL2-dependent mechanisms. Transl Psychiatry 2020; 10:191. [PMID: 32546752 PMCID: PMC7297973 DOI: 10.1038/s41398-020-00876-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/19/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Prenatal stress (PNS) is associated with neuropsychiatric disorders in offspring, including anxiety, depression, and autism spectrum disorders. There is mounting evidence that these behavioral phenotypes have origins in utero. Maternal microbes, inflammation, and serotonergic dysfunction have been implicated as potential mediators of the behavioral consequences of PNS; whether and how these systems interact is unclear. Here, we examine the effects of PNS in utero using late-gestation maternal restraint stress in wild-type (WT), germ-free (GF), and CCL2-/- genetic knock-out (KO) mice. In WT mice, PNS leads to placental and fetal brain inflammation, including an elevation in the chemokine CCL2. This inflammation is largely absent in GF mice, indicating the critical role of maternal microbes in mediating immune processes in utero. Furthermore, PNS in the absence of CCL2 failed to increase pro-inflammatory cytokine IL-6 in the fetal brain. PNS offspring also exhibited deficits in sociability and anxiety-like behavior that were absent in CCL2-/- PNS offspring. Tryptophan and serotonin (5-HT) were elevated in the WT PNS placenta, but not in CCL2-/- and GF animals. Altogether, these findings suggest that a complex interaction between maternal microbes, inflammation, and serotonin metabolism regulates the emergence of behavioral abnormalities following PNS.
Collapse
Affiliation(s)
- Helen J. Chen
- grid.412332.50000 0001 1545 0811Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Adrienne M. Antonson
- grid.412332.50000 0001 1545 0811Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH USA
| | - Therese A. Rajasekera
- grid.412332.50000 0001 1545 0811Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Environmental Health Sciences Division, College of Public Health, The Ohio State University, Columbus, OH USA
| | - Jenna M. Patterson
- grid.412332.50000 0001 1545 0811Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Michael T. Bailey
- grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH USA ,grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, The Research Institute, Nationwide Children’s Hospital, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Tamar L. Gur
- grid.412332.50000 0001 1545 0811Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH USA
| |
Collapse
|
129
|
Czamara D. The Maternal Immunome as a Potential Biomarker for the Child's Neurodevelopmental Trajectory. Biol Psychiatry 2020; 87:868-869. [PMID: 32381155 DOI: 10.1016/j.biopsych.2020.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
130
|
Girchenko P, Lahti-Pulkkinen M, Heinonen K, Reynolds RM, Laivuori H, Lipsanen J, Villa PM, Hämäläinen E, Kajantie E, Lahti J, Räikkönen K. Persistently High Levels of Maternal Antenatal Inflammation Are Associated With and Mediate the Effect of Prenatal Environmental Adversities on Neurodevelopmental Delay in the Offspring. Biol Psychiatry 2020; 87:898-907. [PMID: 31987493 DOI: 10.1016/j.biopsych.2019.12.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Prenatal exposure to environmental adversities, including maternal overweight/obesity, diabetes/hypertensive disorders, or mood/anxiety disorders, increases the risk for adverse neurodevelopmental outcomes in children. However, the underlying biological mechanisms remain elusive. We tested whether maternal antenatal inflammation was associated with the number of neurodevelopmental delay areas in children and whether it mediated the association between exposure to any prenatal environmental adversity and child neurodevelopmental delay. METHODS Mother-child dyads (N = 418) from the PREDO (Prediction and Prevention of Preeclampsia and Intrauterine Growth Restriction) study were followed up to 10.8 years. We analyzed maternal plasma high-sensitivity C-reactive protein and glycoprotein acetyls at 3 consecutive antenatal time points, measured maternal body mass index in early pregnancy, extracted data on diabetes/hypertensive disorders in pregnancy from medical records, and extracted data on mood/anxiety disorders until childbirth from the Care Register for Health Care. To estimate the number of neurodevelopmental delay areas in children across cognitive, motor, and social functioning, we pooled data from the Care Register for Health Care on psychological development disorders with mother-reported Ages and Stages Questionnaire data on developmental milestones. RESULTS Higher levels of maternal high-sensitivity C-reactive protein and glycoprotein acetyls at and across all 3 antenatal time points were associated with 1.30- to 2.36-fold (p values < .02) increased relative risk for higher number of areas of child neurodevelopmental delay. Higher maternal inflammation across the 3 time points also mediated the effect of any prenatal environmental adversity on child neurodevelopmental delay. CONCLUSIONS Higher levels of maternal inflammation, especially when persisting throughout pregnancy, increase a child's risk of neurodevelopmental delay and mediate the effect of prenatal environmental adversity on child neurodevelopmental delay.
Collapse
Affiliation(s)
- Polina Girchenko
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Kati Heinonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rebecca M Reynolds
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, Tampere University Hospital and Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Jari Lipsanen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pia M Villa
- Department of Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eero Kajantie
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; PEDEGO Research Unit, MRC Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Department of Clinical and Molecular Medicine, Norwegian University for Science and Technology, Trondheim, Norway
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
131
|
Davies C, Segre G, Estradé A, Radua J, De Micheli A, Provenzani U, Oliver D, Salazar de Pablo G, Ramella-Cravaro V, Besozzi M, Dazzan P, Miele M, Caputo G, Spallarossa C, Crossland G, Ilyas A, Spada G, Politi P, Murray RM, McGuire P, Fusar-Poli P. Prenatal and perinatal risk and protective factors for psychosis: a systematic review and meta-analysis. Lancet Psychiatry 2020; 7:399-410. [PMID: 32220288 DOI: 10.1016/s2215-0366(20)30057-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/24/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prenatal and perinatal insults are implicated in the aetiopathogenesis of psychotic disorders but the consistency and magnitude of their associations with psychosis have not been updated for nearly two decades. The aim of this systematic review and meta-analysis was to provide a comprehensive and up-to-date synthesis of the evidence on the association between prenatal or perinatal risk and protective factors and psychotic disorders. METHODS In this systematic review and meta-analysis, we searched the Web of Science database for articles published up to July 20, 2019. We identified cohort and case-control studies examining the association (odds ratio [OR]) between prenatal and perinatal factors and any International Classification of Diseases (ICD) or Diagnostic and Statistical Manual of Mental Disorders (DSM) non-organic psychotic disorder with a healthy comparison group. Other inclusion criteria were enough data available to do the analyses, and non-overlapping datasets. We excluded reviews, meta-analyses, abstracts or conference proceedings, and articles with overlapping datasets. Data were extracted according to EQUATOR and PRISMA guidelines. Extracted variables included first author, publication year, study type, sample size, type of psychotic diagnosis (non-affective psychoses or schizophrenia-spectrum disorders, affective psychoses) and diagnostic instrument (DSM or ICD and version), the risk or protective factor, and measure of association (primary outcome). We did random-effects pairwise meta-analyses, Q statistics, I2 index, sensitivity analyses, meta-regressions, and assessed study quality and publication bias. The study protocol was registered at PROSPERO, CRD42017079261. FINDINGS 152 studies relating to 98 risk or protective factors were eligible for analysis. Significant risk factors were: maternal age younger than 20 years (OR 1·17) and 30-34 years (OR 1·05); paternal age younger than 20 years (OR 1·31) and older than 35 years (OR 1·28); any maternal (OR 4·60) or paternal (OR 2·73) psychopathology; maternal psychosis (OR 7·61) and affective disorder (OR 2·26); three or more pregnancies (OR 1·30); herpes simplex 2 (OR 1·35); maternal infections not otherwise specified (NOS; OR 1·27); suboptimal number of antenatal visits (OR 1·83); winter (OR 1·05) and winter to spring (OR 1·05) season of birth in the northern hemisphere; maternal stress NOS (OR 2·40); famine (OR 1·61); any famine or nutritional deficits in pregnancy (OR 1·40); maternal hypertension (OR 1·40); hypoxia (OR 1·63); ruptured (OR 1·86) and premature rupture (OR 2·29) of membranes; polyhydramnios (OR 3·05); definite obstetric complications NOS (OR 1·83); birthweights of less than 2000 g (OR 1·84), less than 2500 g (OR 1·53), or 2500-2999 g (OR 1·23); birth length less than 49 cm (OR 1·17); small for gestational age (OR 1·40); premature birth (OR 1·35), and congenital malformations (OR 2·35). Significant protective factors were maternal ages 20-24 years (OR 0·93) and 25-29 years (OR 0·92), nulliparity (OR 0·91), and birthweights 3500-3999 g (OR 0·90) or more than 4000 g (OR 0·86). The results were corrected for publication biases; sensitivity and meta-regression analyses confirmed the robustness of these findings for most factors. INTERPRETATION Several prenatal and perinatal factors are associated with the later onset of psychosis. The updated knowledge emerging from this study could refine understanding of psychosis pathogenesis, enhance multivariable risk prediction, and inform preventive strategies. FUNDING None.
Collapse
Affiliation(s)
- Cathy Davies
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Giulia Segre
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Andrés Estradé
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Clinical and Health Psychology, Universidad Católica, Montevideo, Uruguay
| | - Joaquim Radua
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Imaging of Mood and Anxiety-Related Disorders (IMARD) group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, Barcelona, Spain; Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Stockholm, Sweden
| | - Andrea De Micheli
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| | - Umberto Provenzani
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Dominic Oliver
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gonzalo Salazar de Pablo
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Institute of Psychiatry and Mental Health, Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, Universidad Complutense, CIBERSAM, Madrid, Spain
| | - Valentina Ramella-Cravaro
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Maria Besozzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Maddalena Miele
- Perinatal Mental Health Service, St Mary's Hospital, Imperial College London and Central North West London NHS Foundation Trust, London, UK
| | - Gianluigi Caputo
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Cecilia Spallarossa
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Georgia Crossland
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Athif Ilyas
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Giulia Spada
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK; Outreach And Support in South London (OASIS) Service, South London and Maudsley NHS Foundation Trust, London, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK; Outreach And Support in South London (OASIS) Service, South London and Maudsley NHS Foundation Trust, London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
| |
Collapse
|
132
|
Yeung EH, Guan W, Zeng X, Salas LA, Mumford SL, de Prado Bert P, van Meel ER, Malmberg A, Sunyer J, Duijts L, Felix JF, Czamara D, Hämäläinen E, Binder EB, Räikkönen K, Lahti J, London SJ, Silver RM, Schisterman EF. Cord blood DNA methylation reflects cord blood C-reactive protein levels but not maternal levels: a longitudinal study and meta-analysis. Clin Epigenetics 2020; 12:60. [PMID: 32354366 PMCID: PMC7193358 DOI: 10.1186/s13148-020-00852-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/15/2020] [Indexed: 02/22/2023] Open
Abstract
Background Prenatal inflammation has been proposed as an important mediating factor in several adverse pregnancy outcomes. C-reactive protein (CRP) is an inflammatory cytokine easily measured in blood. It has clinical value due to its reliability as a biomarker for systemic inflammation and can indicate cellular injury and disease severity. Elevated levels of CRP in adulthood are associated with alterations in DNA methylation. However, no studies have prospectively investigated the relationship between maternal CRP levels and newborn DNA methylation measured by microarray in cord blood with reasonable epigenome-wide coverage. Importantly, the timing of inflammation exposure during pregnancy may also result in different effects. Thus, our objective was to evaluate this prospective association of CRP levels measured during multiple periods of pregnancy and in cord blood at delivery which was available in one cohort (i.e., Effects of Aspirin in Gestation and Reproduction trial), and also to conduct a meta-analysis with available data at one point in pregnancy from three other cohorts from the Pregnancy And Childhood Epigenetics consortium (PACE). Secondarily, the impact of maternal randomization to low dose aspirin prior to pregnancy on methylation was assessed. Results Maternal CRP levels were not associated with newborn DNA methylation regardless of gestational age of measurement (i.e., CRP at approximately 8, 20, and 36 weeks among 358 newborns in EAGeR). There also was no association in the meta-analyses (all p > 0.5) with a larger sample size (n = 1603) from all participating PACE cohorts with available CRP data from first trimester (< 18 weeks gestation). Randomization to aspirin was not associated with DNA methylation. On the other hand, newborn CRP levels were significantly associated with DNA methylation in the EAGeR trial, with 33 CpGs identified (FDR corrected p < 0.05) when both CRP and methylation were measured at the same time point in cord blood. The top 7 CpGs most strongly associated with CRP resided in inflammation and vascular-related genes. Conclusions Maternal CRP levels measured during each trimester were not associated with cord blood DNA methylation. Rather, DNA methylation was associated with CRP levels measured in cord blood, particularly in gene regions predominately associated with angiogenic and inflammatory pathways. Trial registration Clinicaltrials.gov, NCT00467363, Registered April 30, 2007, http://www.clinicaltrials.gov/ct2/show/NCT00467363
Collapse
Affiliation(s)
- Edwina H Yeung
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr, MSC 7004, Bethesda, MD, 20817, USA.
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, A460 Mayo Building, MMC 303, 420 Delaware St. SE, Minneapolis, MN, 55455, USA
| | | | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH, 03766, USA
| | - Sunni L Mumford
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr, MSC 7004, Bethesda, MD, 20817, USA
| | - Paula de Prado Bert
- ISGlobal, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Evelien R van Meel
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Anni Malmberg
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jordi Sunyer
- ISGlobal, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), 08003, Barcelona, Spain
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, Munich, Germany
| | | | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, Munich, Germany.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, USA
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, 27709, USA
| | - Robert M Silver
- University of Utah, Salt Lake City, 50 N Medical Dr, Salt Lake City, UT, 84132, USA
| | - Enrique F Schisterman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr, MSC 7004, Bethesda, MD, 20817, USA
| |
Collapse
|
133
|
Csaba G. Reprogramming of the Immune System by Stress and Faulty Hormonal Imprinting. Clin Ther 2020; 42:983-992. [PMID: 32307123 DOI: 10.1016/j.clinthera.2020.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Hormonal imprinting is taking place perinatally at the first encounter between the developing hormone receptors and their target hormones. However, in this crucial period when the developmental window for physiological imprinting is open, other molecules, such as synthetic hormones and endocrine disruptors can bind to the receptors, leading to faulty imprinting with life-long consequences, especially to the immune system. This review presents the factors of stress and faulty hormonal imprinting that lead to reprogramming of the immune system. METHODS Relevant publications from Pubmed since 1990 were reviewed and synthesized. FINDINGS The developing immune system is rather sensitive to hormonal effects. Faulty hormonal imprinting is able to reprogram the original developmental program present in a given cell, with lifelong consequences, manifested in alteration of hormone binding by receptors, susceptibility to certain (non-infectious) diseases, and triggering of other diseases. As stress mobilizes the hypothalamic-pituitary-adrenal axis if it occurred during gestation or perinatally, it could lead to faulty hormonal imprinting in the immune system, manifested later as allergic and autoimmune diseases or weakness of normal immune defenses. Hormonal imprinting is an epigenetic process and is carried to the offspring without alteration of DNA base sequences. This means that any form of early-life stress alone or in association with hormonal imprinting could be associated with the developmental origin of health and disease (DOHaD). As puberty is also a period of reprogramming, stress or faulty imprinting can change the original (developmental) program, also with life-long consequences. IMPLICATIONS Considering the continuous differentiation of immune cells (from blast-cells) during the whole life, there is a possibility of late-imprinting or stress-activated reprogramming in the immune system at any periods of life, with later pathogenetic consequences.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell, and Immunobiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
134
|
Khambadkone SG, Cordner ZA, Tamashiro KLK. Maternal stressors and the developmental origins of neuropsychiatric risk. Front Neuroendocrinol 2020; 57:100834. [PMID: 32084515 PMCID: PMC7243665 DOI: 10.1016/j.yfrne.2020.100834] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.
Collapse
Affiliation(s)
- Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
135
|
Lautarescu A, Pecheva D, Nosarti C, Nihouarn J, Zhang H, Victor S, Craig M, Edwards AD, Counsell SJ. Maternal Prenatal Stress Is Associated With Altered Uncinate Fasciculus Microstructure in Premature Neonates. Biol Psychiatry 2020; 87:559-569. [PMID: 31604519 PMCID: PMC7016501 DOI: 10.1016/j.biopsych.2019.08.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Maternal prenatal stress exposure (PNSE) increases risk for adverse psychiatric and behavioral outcomes in offspring. The biological basis for this elevated risk is poorly understood but may involve alterations to the neurodevelopmental trajectory of white matter tracts within the limbic system, particularly the uncinate fasciculus. Additionally, preterm birth is associated with both impaired white matter development and adverse developmental outcomes. In this study we hypothesized that higher maternal PNSE was associated with altered uncinate fasciculus microstructure in offspring. METHODS In this study, 251 preterm infants (132 male, 119 female) (median gestational age = 30.29 weeks [range, 23.57-32.86 weeks]) underwent brain magnetic resonance imaging including diffusion-weighted imaging around term-equivalent age (median = 42.43 weeks [range, 37.86-45.71 weeks]). Measures of white matter microstructure were calculated for the uncinate fasciculus and the inferior longitudinal fasciculus, a control tract that we hypothesized was not associated with maternal PNSE. Multiple regressions were used to investigate the relationship among maternal trait anxiety scores, stressful life events, and white matter microstructure indices in the neonatal brain. RESULTS Adjusting for gestational age at birth, postmenstrual age at scan, maternal age, socioeconomic status, sex, and number of days on parenteral nutrition, higher stressful life events scores were associated with higher axial diffusivity (β = .177, q = .007), radial diffusivity (β = .133, q = .026), and mean diffusivity (β = .149, q = .012) in the left uncinate fasciculus, and higher axial diffusivity (β = .142, q = .026) in the right uncinate fasciculus. CONCLUSIONS These findings suggest that PNSE is associated with altered development of specific frontolimbic pathways in preterm neonates as early as term-equivalent age.
Collapse
Affiliation(s)
- Alexandra Lautarescu
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | - Diliana Pecheva
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Chiara Nosarti
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Julie Nihouarn
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Hui Zhang
- Department of Computer Science and Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Suresh Victor
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Michael Craig
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom,National Female Hormone Clinic, South London and Maudsley National Health Service Foundation Trust, London, United Kingdom
| | - A. David Edwards
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Serena J. Counsell
- Department of Perinatal Imaging and Health, Centre for Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
136
|
Agustini B, Berk M. The embryology of psychosis. Lancet Psychiatry 2020; 7:219-220. [PMID: 32035029 DOI: 10.1016/s2215-0366(20)30042-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Bruno Agustini
- Deakin University, Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Barwon Health, Geelong, VIC 3220, Australia.
| | - Michael Berk
- Deakin University, Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Barwon Health, Geelong, VIC 3220, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Melbourne, VIC, Australia; Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia; Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
137
|
Phua DY, Kee MZL, Meaney MJ. Positive Maternal Mental Health, Parenting, and Child Development. Biol Psychiatry 2020; 87:328-337. [PMID: 31839213 DOI: 10.1016/j.biopsych.2019.09.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/23/2022]
Abstract
While maternal mental health is an important influence on child development, the existing literature focuses primarily on negative aspects of maternal mental health, particularly symptoms of depression, anxiety, or states of distress. We provide a review of the evidence on the potential importance of positive mental health for both mother and child. The evidence suggests that positive mental health is a distinct construct that is associated with improved birth outcomes and potentially with specific forms of parenting that promote both academic achievement and socioemotional function. We review studies that provide a plausible biological basis for the link between positive mental health and parenting, focusing on oxytocin-dopamine interactions. We caution that the evidence is largely preliminary and suggest directions for future research, noting the importance of identifying the operative dimensions of positive maternal mental health in relation to specific outcomes. We suggest that the inclusion of positive maternal mental health provides the potential for a more comprehensive understanding of parental influences on child development.
Collapse
Affiliation(s)
- Desiree Y Phua
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Michelle Z L Kee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
138
|
Wu Y, Lu YC, Jacobs M, Pradhan S, Kapse K, Zhao L, Niforatos-Andescavage N, Vezina G, du Plessis AJ, Limperopoulos C. Association of Prenatal Maternal Psychological Distress With Fetal Brain Growth, Metabolism, and Cortical Maturation. JAMA Netw Open 2020; 3:e1919940. [PMID: 31995213 PMCID: PMC6991285 DOI: 10.1001/jamanetworkopen.2019.19940] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPORTANCE Prenatal maternal stress is increasingly associated with adverse outcomes in pregnant women and their offspring. However, the association between maternal stress and human fetal brain growth and metabolism is unknown. OBJECTIVE To identify the association between prenatal maternal psychological distress and fetal brain growth, cortical maturation, and biochemical development using advanced 3-dimensional volumetric magnetic resonance imaging (MRI) and proton magnetic resonance spectroscopy (1H-MRS). DESIGN, SETTING, AND PARTICIPANTS This cohort study prospectively recruited pregnant women from low-risk obstetric clinics in Washington, DC, from January 1, 2016, to April 17, 2019. Participants were healthy volunteers with a normal prenatal medical history, no chronic or pregnancy-induced physical or mental illnesses, and normal results on fetal ultrasonography and biometry studies. Fetal brain MRI studies were performed at 2 time points between 24 and 40 weeks' gestation. EXPOSURES Prenatal maternal stress, anxiety, and depression. MAIN OUTCOMES AND MEASURES Volumes of fetal total brain, cortical gray matter, white matter, deep gray matter, cerebellum, brainstem, and hippocampus were measured from 3-dimensional reconstructed T2-weighted MRI scans. Cortical folding measurements included local gyrification index, sulcal depth, and curvedness. Fetal brain N-acetylaspartate, creatine, and choline levels were quantified using 1H-MRS. Maternal stress, depression, and anxiety were measured with the Perceived Stress Scale (PSS), Edinburgh Postnatal Depression Scale (EPDS), Spielberger State Anxiety Inventory (SSAI), and Spielberger Trait Anxiety Inventory (STAI). RESULTS A total of 193 MRI studies were performed in 119 pregnant women (67 [56%] carrying male fetuses and 52 [44%], female fetuses; maternal mean [SD] age, 34.46 [5.95] years) between 24 and 40 gestational weeks. All women were high school graduates, 99 (83%) were college graduates, and 100 (84%) reported professional employment. Thirty-two women (27%) had positive scores for stress, 31 (26%) for anxiety, and 13 (11%) for depression. Maternal trait anxiety was associated with smaller fetal left hippocampal volume (STAI score: -0.002 cm3; 95% CI, -0.003 to -0.0008 cm3; P = .004). Maternal anxiety and stress were associated with increased fetal cortical gyrification in the frontal lobe (β for SSAI score: 0.004 [95% CI, 0.001-0.006; P = .002]; β for STAI score: 0.004 [95% CI, 0.002-0.006; P < .001]; β for PSS score: 0.005 [95% CI, 0.001-0.008; P = .005]) and temporal lobe (β for SSAI score: 0.004 [95% CI, 0.001-0.007; P = .004]; β for STAI score: 0.004 [95% CI, 0.0008-0.006; P = .01]). Elevated maternal depression was associated with decreased creatine (EPDS score: -0.04; 95% CI, -0.06 to -0.02; P = .005) and choline (EPDS score: -0.03; 95% CI, -0.05 to -0.01; P = .02) levels in the fetal brain. CONCLUSIONS AND RELEVANCE This study found that the prevalence of maternal psychological distress in healthy, well-educated, and employed pregnant women was high, underappreciated, and associated with impaired fetal brain biochemistry and hippocampal growth as well as accelerated cortical folding. These findings appear to support the need for routine mental health surveillance for all pregnant women and targeted interventions in women with elevated psychological distress.
Collapse
Affiliation(s)
- Yao Wu
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
| | - Yuan-Chiao Lu
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
| | - Marni Jacobs
- Department of Biostatistics and Study Methodology, Children’s Research Institute, Children’s National Hospital, Washington, DC
| | - Subechhya Pradhan
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
| | - Kushal Kapse
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
| | - Li Zhao
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
| | | | - Gilbert Vezina
- Department of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, DC
| | | | - Catherine Limperopoulos
- Center for the Developing Brain, Children’s National Hospital, Washington, DC
- Department of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, DC
| |
Collapse
|
139
|
Duman EA, Atesyakar N, Ecevitoglu A. Multilevel Impact of Prenatal Risk and Protective Factors on Stress Biology and Infant Development: Study protocol of BABIP prospective birth cohort from Turkey. Brain Behav Immun Health 2020; 1:100005. [PMID: 38377425 PMCID: PMC8474236 DOI: 10.1016/j.bbih.2019.100005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 11/19/2022] Open
Abstract
Prenatal environment has long-lasting effects on offspring development and health. Research on prenatal stress identified various mechanisms of these effects, from changes in epigenetic and gene expression profiles to Maternal-Placental-Fetal (MPF) stress biology. There is also evidence for the role of additional risk and protective factors influencing the impact of prenatal stress on maternal and infant outcomes. Considering these findings, we present the study protocol of BABIP, a prospective birth cohort from Turkey. The aim of the project is to investigate the effect of prenatal stress on MPF stress biology (i.e. neuroendocrine, immune and metabolic systems), differential DNA methylation and gene expression patterns, and infant birth and developmental outcomes. We are recruiting 150 pregnant women and their babies for a longitudinal project with 4 time points: 20-24 (T1) and 30-34 (T2) weeks of pregnancy, and 1-month (T3) and 4-months (T4) after giving birth. Maternal early and prenatal environment (prenatal stress, early life stress, psychosocial resources, and health-related behaviors) are assessed during pregnancy with MPF stress biology, DNA methylation and gene expression measures. Infant birth outcomes, DNA methylation and development are assessed postpartum. BABIP is the first prospective birth cohort from Turkey with extensive measures on prenatal environment and health. Through investigating the multilevel impact of prenatal stress and related risk and protective factors during and after pregnancy, BABIP will contribute to our understanding of the mechanisms by which prenatal environment influences infant development and health. Being the first such cohort from Turkey, it may also allow identification of prenatal risk and protective factors specific to the context and population in Turkey.
Collapse
Affiliation(s)
- Elif Aysimi Duman
- Department of Psychology, Bogazici University, Istanbul, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Istanbul, Turkey
| | - Nilay Atesyakar
- Department of Psychology, Bogazici University, Istanbul, Turkey
| | - Alev Ecevitoglu
- Department of Psychology, Bogazici University, Istanbul, Turkey
| |
Collapse
|
140
|
Watkins JM, von Chamier M, Brown MB, Reyes L, Hayward LF. Prenatal infection with Mycoplasma pulmonis in rats exaggerates the angiotensin II pressor response in adult offspring. Am J Physiol Regul Integr Comp Physiol 2019; 318:R338-R350. [PMID: 31850818 DOI: 10.1152/ajpregu.00194.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure to different stressors in utero is linked to adult diseases such as obesity and hypertension. In this study, the impact of prenatal infection (PNI) on adult body weight and cardiovascular function was evaluated using a naturally occurring rodent pathogen, Mycoplasma pulmonis (MP). Pregnant Sprague-Dawley rats were infected with MP on gestational day 14 and gave birth naturally. Adult PNI offspring weighed more than controls, but resting mean arterial pressure (MAP) was unchanged. Subcutaneous injection of angiotensin II (10 μg/kg) elicited a rise in MAP that was greater in both male and female PNI offspring compared with controls (P < 0.03). The accompanying reflex bradycardia was similar to the controls, suggesting that PNI induced baroreflex dysfunction. Subcutaneous nicotine administration, a potent cardiorespiratory stimulus, also elicited a transient rise in MAP that was generally greater in the PNI group, but the change in MAP from baseline was only significant in the PNI females compared with controls (P < 0.03). Elevated body weight and cardiovascular reactivity in the PNI offspring was associated with an increase in the ratio of hypothalamic corticotrophin-releasing hormone receptors type 1 to type 2 gene expression in both sexes compared with controls. These findings support previous studies demonstrating that PNI induces alterations in cardiovascular function and body weight. Yet, unlike previous studies utilizing other models of PNI (e.g., endotoxin), MP PNI did not induce resting hypertension. Thus, our study provides a foundation for future studies evaluating the cardiovascular risks of offspring exposed to microbial challenges in utero.
Collapse
Affiliation(s)
- J M Watkins
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - M von Chamier
- Department of Infectious Disease and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - M B Brown
- Department of Infectious Disease and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - L Reyes
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - L F Hayward
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
141
|
Meyer U. Neurodevelopmental Resilience and Susceptibility to Maternal Immune Activation. Trends Neurosci 2019; 42:793-806. [DOI: 10.1016/j.tins.2019.08.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/05/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022]
|
142
|
Lipner E, Murphy SK, Ellman LM. Prenatal Maternal Stress and the Cascade of Risk to Schizophrenia Spectrum Disorders in Offspring. Curr Psychiatry Rep 2019; 21:99. [PMID: 31522269 PMCID: PMC7043262 DOI: 10.1007/s11920-019-1085-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Disruptions in fetal development (via genetic and environmental pathways) have been consistently associated with risk for schizophrenia in a variety of studies. Although multiple obstetric complications (OCs) have been linked to schizophrenia, this review will discuss emerging evidence supporting the role of prenatal maternal stress (PNMS) in the etiology of schizophrenia spectrum disorders (SSD). In addition, findings linking PNMS to intermediate phenotypes of the disorder, such as OCs and premorbid cognitive, behavioral, and motor deficits, will be reviewed. Maternal immune and endocrine dysregulation will also be explored as potential mechanisms by which PNMS confers risk for SSD. RECENT FINDINGS PNMS has been linked to offspring SSD; however, findings are mixed due to inconsistent and retrospective assessments of PNMS and lack of specificity about SSD outcomes. PNMS is also associated with various intermediate phenotypes of SSD (e.g., prenatal infection/inflammation, decreased fetal growth, hypoxia-related OCs). Recent studies continue to elucidate the impact of PNMS while considering the moderating roles of fetal sex and stress timing, but it is still unclear which aspects of PNMS (e.g., type, timing) confer risk for SSD specifically. PNMS increases risk for SSD, but only in a small portion of fetuses exposed to PNMS. Fetal sex, genetics, and other environmental factors, as well as additional pre- and postnatal insults, likely contribute to the PNMS-SSD association. Longitudinal birth cohort studies are needed to prospectively illuminate the mechanisms that account for the variability in outcomes following PNMS.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA
| | - Shannon K Murphy
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA, 19106, USA.
| |
Collapse
|
143
|
McLean MA, Cobham VE, Simcock G, Kildea S, King S. Toddler Temperament Mediates the Effect of Prenatal Maternal Stress on Childhood Anxiety Symptomatology: The QF2011 Queensland Flood Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16111998. [PMID: 31195616 PMCID: PMC6603961 DOI: 10.3390/ijerph16111998] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022]
Abstract
It is not known whether alterations to temperamental characteristics associated with prenatal maternal stress (PNMS) exposure account for the development of childhood anxiety symptomatology (internalizing behaviors and anxiety symptoms). The QF2011 Queensland flood study examined whether (1) toddler temperamental characteristics explained the association between PNMS exposure and childhood anxiety symptomatology; and (2) whether effects were dependent upon child sex or the timing of gestational exposure to PNMS. We investigated the effects of various aspects of flood-related stress in pregnancy (objective hardship, cognitive appraisal, subjective distress) on maternal report of 16-month toddler temperament (attentional control, shy-inhibition, negative reactivity), 4-year maternal-reported childhood anxiety symptomatology (internalizing and anxiety symptoms; N = 104), and teacher reports of internalizing behaviors (N = 77). Severity of maternal objective hardship during pregnancy and shy-inhibited behaviors were uniquely associated with 4-year child anxiety symptoms. Mediation analyses found that higher levels of 16-month negative reactivity accounted, in part, for the relationship between increased maternal objective flood-related hardship and greater internalizing behaviors (maternal but not teacher report). Neither child sex nor gestational timing of exposure moderated the hypothesized mediations. Our findings highlight several pathways through which varying aspects of disaster-related PNMS may influence early childhood anxiety symptomatology.
Collapse
Affiliation(s)
- Mia A McLean
- Mater Research Institute-University of Queensland, Brisbane, QLD 4072, Australia.
- School of Psychology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Vanessa E Cobham
- Mater Research Institute-University of Queensland, Brisbane, QLD 4072, Australia.
- School of Psychology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Gabrielle Simcock
- Mater Research Institute-University of Queensland, Brisbane, QLD 4072, Australia.
- Sunshine Coast Mind and Neuroscience Thompson Institute, University of Sunshine Coast, Sippy Downs, QLD 4556, Australia.
| | - Sue Kildea
- Mater Research Institute-University of Queensland, Brisbane, QLD 4072, Australia.
- School of Nursing, Midwifery, and Social Work, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Suzanne King
- Schizophrenia and Neurodevelopmental Disorders Research, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada.
| |
Collapse
|
144
|
McEwen BS. Prenatal Programming of Neuropsychiatric Disorders: An Epigenetic Perspective Across the Lifespan. Biol Psychiatry 2019; 85:91-93. [PMID: 30573051 DOI: 10.1016/j.biopsych.2018.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York.
| |
Collapse
|