1
|
Wen Y, Zhou J, Yu H, Wu Z, Peng R, Xu C, Shi X, Jiang M, Yuan H, Feng S. Esketamine ameliorates prenatal stress-induced postpartum depression and sex-related behavioral differences in adolescent progeny. Neuropharmacology 2025; 269:110354. [PMID: 39947391 DOI: 10.1016/j.neuropharm.2025.110354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Prenatal stress leads to postpartum depression and is associated with developmental issues in offspring. Esketamine quickly and effectively prevents or treats postpartum depression. However, the long-term effects of esketamine on progeny development are unknown. METHODS CRS during pregnancy was used to establish the postpartum depression animal model. After confirming pregnancy, all mice were randomly divided into three groups: pregnant control group, pregnant restraint group, and pregnant restraint + esketamine group. Mice in the restraint + esketamine group received esketamine intraperitoneal injection on postpartum days 1-5. Behavioral tests were performed on maternal mice five days after delivery. Another cohort of mice was used to test the effects of esketamine on the behavior of offspring mice. The levels of ACTH and CORT were measured in offspring mice in response to acute restraint stress by ELISA. RESULTS We found that prenatal CRS induced postpartum depression-like behaviors in maternal mice and sex-related differential behaviors in adolescent offspring. Female offspring exhibited depression-like behaviors, and male offspring showed memory deficits. Esketamine improved postpartum depression-like behaviors in maternal mice and behavioral changes in teenage offspring. Prenatal CRS led to hyperresponsiveness of ACTH and CORT to acute restraint stress in adolescent offspring. Compared with the offspring in the control group, the restraint group increased secretion of ACTH and CORT during acute restraint stress. CONCLUSION Prenatal CRS led to postpartum depression-like behaviors in maternal mice and sex-related differential behaviors in adolescent offspring. Esketamine effectively improves postpartum depression-like behaviors in maternal mice and behavioral changes in adolescent offspring.
Collapse
Affiliation(s)
- Yazhou Wen
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Jin Zhou
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Huiling Yu
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Zixin Wu
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Rui Peng
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Chenyang Xu
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Xueduo Shi
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ming Jiang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China.
| | - Hongmei Yuan
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China.
| | - Shanwu Feng
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China.
| |
Collapse
|
2
|
Muszyński J, Bienert A, Elsorady RW, Rybakowski F. New pharmacological approaches in the treatment of schizophrenia. Pharmacol Rep 2025:10.1007/s43440-025-00722-9. [PMID: 40198498 DOI: 10.1007/s43440-025-00722-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Schizophrenia is a primary health concern, imposing a significant burden on both patients and healthcare systems globally. It is a disease with a complex etiology in which both genetic and environmental factors are involved. Despite numerous studies, the mechanism of its origin is still not fully understood. The hypotheses are synaptic, serotonergic, muscarinic, dopaminergic, microRNA-related, and neurodegenerative theories. Treatment to date is mainly based on antipsychotic drugs that act on the dopaminergic system. Although they are effective in reducing positive symptoms, their effect on negative and cognitive symptoms is limited, and their use is often associated with numerous side effects. A breakthrough in the treatment of schizophrenia came with the approval of the first drug with a non-dopaminergic mechanism of action, which opens up new therapeutic possibilities. As a result, there is intensive research into innovative substances that could increase the effectiveness of treatment and improve the quality of life of patients. In this review, we present the current state of knowledge about schizophrenia, its prevalence, risk factors, and its impact on patients' functioning. We pay special attention to new therapeutic directions, including drugs that affect systems other than the dopaminergic one, which could open up new prospects for treating the condition.
Collapse
Affiliation(s)
- Józef Muszyński
- Dr. Jan Jonston Regional Multispecialty Hospital in Leszno, Leszno, Poland
| | - Agnieszka Bienert
- Department of Pharmacology, Poznan University of Medical Sciences, Rokietnicka 3, Poznań, 60-806, Poland.
| | - Rasha Wafaie Elsorady
- Head of Clinical Pharmacy Departments at Alexandria University Hospitals, Alexandria University, Alexandria, 21523, Egypt
| | - Filip Rybakowski
- Head of Adult Psychiatry Clinic, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Yang H, Sun W, Li J, Zhang X. Epigenetics factors in schizophrenia: future directions for etiologic and therapeutic study approaches. Ann Gen Psychiatry 2025; 24:21. [PMID: 40186258 PMCID: PMC11969811 DOI: 10.1186/s12991-025-00557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/14/2025] [Indexed: 04/07/2025] Open
Abstract
Schizophrenia is a complex, heterogeneous, and highly disabling severe mental disorder whose pathogenesis has not yet been fully elucidated. Epigenetics, as a bridge between genetic and environmental factors, plays an important role in the pathophysiology of schizophrenia. Over the past decade, epigenetic-wide association studies have rapidly become an important branch of psychiatric research, especially in deciphering the molecular mechanisms of schizophrenia. This review systematically analyzes recent advances in epigenome-wide association studies (EWAS) of schizophrenia, focusing on technological developments. We synthesize findings from large-scale EWAS alongside emerging evidence on DNA methylation patterns, histone modifications, and regulatory networks, emphasizing their roles in disease mechanisms and treatment responses. In addition, this review provides a prospective outlook, evaluating the impact that technological developments may have on future studies of schizophrenia. With the continuous advancement of high-throughput sequencing technology and the increasing maturity of big data analysis methods, epigenetics is expected to have a significant impact on the early diagnosis, prognosis assessment and even personalized treatment of schizophrenia.
Collapse
Affiliation(s)
- Haidong Yang
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, People's Republic of China
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, People's Republic of China
| | - Wenxi Sun
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, People's Republic of China
| | - Jin Li
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, People's Republic of China
| | - Xiaobin Zhang
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, People's Republic of China.
| |
Collapse
|
4
|
Arakelyan NA, Kupriyanova DA, Vasilevska J, Rogaev EI. Sexual dimorphism in immunity and longevity among the oldest old. Front Immunol 2025; 16:1525948. [PMID: 40034689 PMCID: PMC11872714 DOI: 10.3389/fimmu.2025.1525948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Human longevity is a sex-biased process in which sex chromosomes and sex-specific immunity may play a crucial role in the health and lifespan disparities between men and women. Generally, women have a higher life expectancy than men, exhibiting lower infection rates for a broad range of pathogens, which results in a higher prevalence of female centenarians compared to males. Investigation of the immunological changes that occur during the process of healthy aging, while taking into account the differences between sexes, can significantly enhance our understanding of the mechanisms that underlie longevity. In this review, we aim to summarize the current knowledge on sexual dimorphism in the human immune system and gut microbiome during aging, with a particular focus on centenarians, based exclusively on human data.
Collapse
Affiliation(s)
- Nelli A. Arakelyan
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Daria A. Kupriyanova
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Jelena Vasilevska
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
5
|
Maurer SV, Hing BWQ, Lussier S, Radhakrishna S, Davis JLB, Abbott PW, Michaelson JJ, Stevens HE. Prenatal stress alters mouse offspring dorsal striatal development and placental function in sex-specific ways. J Psychiatr Res 2025; 182:149-160. [PMID: 39809011 PMCID: PMC11959308 DOI: 10.1016/j.jpsychires.2024.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Prenatal stress is a risk factor for neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD). However, how early stress modification of brain development contributes to this pathophysiology is poorly understood. Ventral forebrain regions such as dorsal striatum are of particular interest: dorsal striatum modulates movement and cognition, is altered in NDDs, and has a primarily GABAergic population. Here, we examine effects of prenatal stress on adult movement, cognition, and dorsal striatum neurobiology in mice using striatal-dependent behavioral assays, immunohistochemistry, embryonic ventral forebrain transcriptomics, and placental transcriptomics. We found prenatal stress affected adult procedural, habit, and reversal learning in sex-specific ways. Stress also increased adult dorsal striatal GABAergic neurons - an effect largely driven by males. We sought to examine the developmental origins of these adult brain changes. We found similar sex-specific dorsal striatal cellular changes in earlier points of development. The dorsal striatum primordium--embryonic ventral forebrain-showed that prenatal stress increased DNA replication and cell cycle pathways in male but not female transcriptomics and cellular biology. Unique signatures may have arisen from male-female placental differences. Stress-induced placental transcriptomics showed upregulated morphogenesis pathways in males while females downregulated morphogenic, hormonal, and cellular response pathways. Our findings suggest that prenatal stress could affect placenta function and also alter the GABAergic population of dorsal striatum differentially between the sexes.
Collapse
Affiliation(s)
- Sara V Maurer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Benjamin W Q Hing
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Stephanie Lussier
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SL is now with Moderna, USA
| | - Sreya Radhakrishna
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SR is now at Albert Einstein College of Medicine, USA
| | - Jada L B Davis
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Parker W Abbott
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
6
|
Murphy SK, Pike MR, Lipner E, Maxwell SD, Cohn BA, Cirillo P, Krigbaum NY, Breen EC, Ellman LM. Contributions of maternal prenatal infection and antibiotic exposure to offspring infection and risk for allergic respiratory conditions through age 5. Brain Behav Immun Health 2024; 42:100892. [PMID: 39512604 PMCID: PMC11541876 DOI: 10.1016/j.bbih.2024.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/15/2024] Open
Abstract
Objectives To determine if maternal prenatal infection increases risk of offspring postnatal infections through age 5 or diagnosis of respiratory allergy at age 5, independent of prenatal/postnatal antibiotic exposure. To evaluate if frequency of offspring infections mediates an association between prenatal infection and respiratory allergy at age 5. Study design Secondary data analyses were performed from the Child Health and Development Studies (CHDS), a prospective, longitudinal birth cohort that enrolled pregnant women from 1959 to 1966 (N = 19,044 live births). The sample included a subset of mother-offspring dyads (n = 2062) with abstracted medical record data from the prenatal period through age 5 that included information on antibiotic use, infection, and offspring respiratory allergy. Results Second trimester maternal infection was associated with an increased risk of offspring infection (IRR = 1.23; 95% CI = 1.09-1.39; p = 0.001). No significant direct associations were detected between prenatal infection and diagnosis of offspring respiratory allergy. Offspring infection (OR = 1.17; 95% CI = 1.13-1.20; p < 0.001) and antibiotic exposure (OR = 1.28; 95% CI = 1.22-1.33; p < 0.001) were significantly associated with a diagnosis of offspring respiratory allergy. Respiratory allergy diagnosis risk was greater with increasing offspring infection exposure and antibiotics. There was a significant indirect effect of second trimester maternal infection on offspring respiratory allergy, due to infections and not antibiotic use, via offspring infection, indicating a partially mediated effect. Conclusion Prenatal maternal infection may contribute to increase risk for early childhood infections, which in turn, may increase risk for allergic conditions.
Collapse
Affiliation(s)
- Shannon K. Murphy
- Temple University, Department of Psychology & Neuroscience, Philadelphia, PA, USA
| | - Madeline R. Pike
- Temple University, Department of Psychology & Neuroscience, Philadelphia, PA, USA
| | - Emily Lipner
- Temple University, Department of Psychology & Neuroscience, Philadelphia, PA, USA
| | - Seth D. Maxwell
- Temple University, Department of Psychology & Neuroscience, Philadelphia, PA, USA
| | - Barbara A. Cohn
- Child Health and Development Studies, Public Health Institute, Oakland, CA, USA
| | - Piera Cirillo
- Child Health and Development Studies, Public Health Institute, Oakland, CA, USA
| | | | - Elizabeth C. Breen
- Cousins Center for Psychoneuroimmunology, Dept. of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, USA
| | - Lauren M. Ellman
- Temple University, Department of Psychology & Neuroscience, Philadelphia, PA, USA
| |
Collapse
|
7
|
Abrishamcar S, Zhuang BC, Thomas M, Gladish N, MacIsaac JL, Jones MJ, Simons E, Moraes TJ, Mandhane PJ, Brook JR, Subbarao P, Turvey SE, Chen E, Miller GE, Kobor MS, Hüls A. Association between maternal perinatal stress and depression and infant DNA methylation in the first year of life. Transl Psychiatry 2024; 14:445. [PMID: 39438450 PMCID: PMC11496819 DOI: 10.1038/s41398-024-03148-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Maternal stress and depression during pregnancy and the first year of the infant's life affect a large percentage of mothers. Maternal stress and depression have been associated with adverse fetal and childhood outcomes as well as differential child DNA methylation (DNAm). However, the biological mechanisms connecting maternal stress and depression to poor health outcomes in children are still largely unknown. Here we aim to determine whether prenatal stress and depression are associated with differences in cord blood mononuclear cell DNAm (CBMC-DNAm) in newborns (n = 119) and whether postnatal stress and depression are associated with differences in peripheral blood mononuclear cell DNAm (PBMC-DNAm) in children of 12 months of age (n = 113) from the Canadian Healthy Infant Longitudinal Development (CHILD) cohort. Stress was measured using the 10-item Perceived Stress Scale (PSS) and depression was measured using the 20-item Center for Epidemiologic Studies Depression Questionnaire (CESD). Both stress and depression were measured longitudinally at 18 weeks and 36 weeks of pregnancy and six months and 12 months postpartum. We conducted epigenome-wide association studies (EWAS) using robust linear regression followed by a sensitivity analysis in which we bias-adjusted for inflation and unmeasured confounding using the bacon and cate methods. To quantify the cumulative effect of maternal stress and depression, we created composite prenatal and postnatal adversity scores. We identified a significant association between prenatal stress and differential CBMC-DNAm at 8 CpG sites and between prenatal depression and differential CBMC-DNAm at 2 CpG sites. Additionally, we identified a significant association between postnatal stress and differential PBMC-DNAm at 8 CpG sites and between postnatal depression and differential PBMC-DNAm at 11 CpG sites. Using our composite scores, we further identified 2 CpG sites significantly associated with prenatal adversity and 7 CpG sites significantly associated with postnatal adversity. Several of the associated genes, including PLAGL1, HYMAI, BRD2, and ERC2 have been implicated in adverse fetal outcomes and neuropsychiatric disorders. These data further support the finding that differential DNAm may play a role in the relationship between maternal mental health and child health.
Collapse
Affiliation(s)
- Sarina Abrishamcar
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Beryl C Zhuang
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Edwin S.H. Leong Centre for Healthy Aging, Vancouver, BC, Canada
| | - Mara Thomas
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Nicole Gladish
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Edwin S.H. Leong Centre for Healthy Aging, Vancouver, BC, Canada
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Julia L MacIsaac
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Edwin S.H. Leong Centre for Healthy Aging, Vancouver, BC, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Elinor Simons
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Theo J Moraes
- Division of Respiratory Medicine, Department of Pediatrics, Hospital for Sick Children & Research Institute, Toronto, ON, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Padmaja Subbarao
- Division of Respiratory Medicine, Department of Pediatrics, Hospital for Sick Children & Research Institute, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Edith Chen
- Department of Psychology, Northwestern University, Evanston, IL, USA
- Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Gregory E Miller
- Department of Psychology, Northwestern University, Evanston, IL, USA
- Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Edwin S.H. Leong Centre for Healthy Aging, Vancouver, BC, Canada.
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
8
|
Ortiz-Valladares M, Gonzalez-Perez O, Pedraza-Medina R. Bridging the gap: Prenatal nutrition, myelination, and schizophrenia etiopathogenesis. Neuroscience 2024; 558:58-69. [PMID: 39159841 DOI: 10.1016/j.neuroscience.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
Schizophrenia (SZ) is a complex mental illness characterized by disturbances in thinking, emotionality, and behavior, significantly impacting the quality of life for individuals affected and those around them. The etiology of SZ involves intricate interactions between genetic and environmental factors, although the precise mechanisms remain incompletely understood. Genetic predisposition, neurotransmitter dysregulation (particularly involving dopamine and serotonin), and structural brain abnormalities, including impaired prefrontal cortex function, have been implicated in SZ development. However, increasing evidence reveals the role of environmental factors, such as nutrition, during critical periods like pregnancy and lactation. Epidemiological studies suggest that early malnutrition significantly increases the risk of SZ symptoms manifesting in late adolescence, a crucial period coinciding with peak myelination and brain maturation. Prenatal undernutrition may disrupt myelin formation, rendering individuals more susceptible to SZ pathology. This review explores the potential relationship between prenatal undernutrition, myelin alterations, and susceptibility to SZ. By delineating the etiopathogenesis, examining genetic and environmental factors associated with SZ, and reviewing the relationship between SZ and myelination disorders, alongside the impact of malnutrition on myelination, we aim to examine how malnutrition might be linked to SZ by altering myelination processes, which contribute to increasing the understanding of SZ etiology and help identify targets for intervention and management.
Collapse
Affiliation(s)
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040. México
| | - Ricardo Pedraza-Medina
- Medical Science Postgraduate Program, School of Medicine, University of Colima, Colima 28040. México
| |
Collapse
|
9
|
Shaaban M, Shepelak ZD, Stanford JB, Silver RM, Mumford SL, Schisterman EF, Hinkle SN, Nkoy FL, Theilen L, Page J, Woo JG, Brown BH, Varner MW, Schliep KC. Low-dose aspirin, maternal cardiometabolic health, and offspring respiratory health 9 to 14 years after delivery: Findings from the EAGeR Follow-up Study. Paediatr Perinat Epidemiol 2024; 38:570-580. [PMID: 38886184 PMCID: PMC11427166 DOI: 10.1111/ppe.13097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Accumulating evidence shows that peri-conceptional and in-utero exposures have lifetime health impacts for mothers and their offspring. OBJECTIVES We conducted a Follow-Up Study of the Effects of Aspirin in Gestation and Reproduction (EAGeR) trial with two objectives. First, we determined if women who enrolled at the Utah site (N = 1001) of the EAGeR trial (2007-2011, N = 1228) could successfully be contacted and agree to complete an online questionnaire on their reproductive, cardio-metabolic, and offspring respiratory health 9-14 years after original enrollment. Second, we evaluated if maternal exposure to low-dose aspirin (LDA) during pregnancy was associated with maternal cardio-metabolic health and offspring respiratory health. METHODS The original EAGeR study population included women, 18-40 years of age, who had 1-2 prior pregnancy losses, and who were trying to become pregnant. At follow-up (2020-2021), participants from the Utah cohort completed a 13-item online questionnaire on reproductive and cardio-metabolic health, and those who had a live birth during EAGeR additionally completed a 7-item questionnaire on the index child's respiratory health. Primary maternal outcomes included hypertension and hypercholesterolemia; primary offspring outcomes included wheezing and asthma. RESULTS Sixty-eight percent (n = 678) of participants enrolled in the follow-up study, with 10% and 15% reporting maternal hypertension and hypercholesterolemia, respectively; and 18% and 10% reporting offspring wheezing and asthma. We found no association between maternal LDA exposure and hypertension (risk difference [RD] -0.001, 95% confidence interval [CI] -0.05, 0.04) or hypercholesterolemia (RD -0.01, 95% CI -0.06, 0.05) at 9-14 years follow-up. Maternal LDA exposure was not associated with offspring wheezing (RD -0.002, 95% CI -0.08, 0.08) or asthma (RD 0.13, 95% CI 0.11, 0.37) at follow-up. Findings remained robust after considering potential confounding and selection bias. CONCLUSIONS We observed no association between LDA exposure during pregnancy and maternal cardiometabolic or offspring respiratory health.
Collapse
Affiliation(s)
- May Shaaban
- Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | - Zachary D Shepelak
- Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | - Joseph B Stanford
- Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah Health, Salt Lake City, Utah, USA
| | - Sunni L Mumford
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Enrique F Schisterman
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefanie N Hinkle
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Flory L Nkoy
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah, USA
| | - Lauren Theilen
- Department of Obstetrics and Gynecology, University of Utah Health, Salt Lake City, Utah, USA
| | - Jessica Page
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Jessica G Woo
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Benjamin H Brown
- Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | - Michael W Varner
- Department of Obstetrics and Gynecology, University of Utah Health, Salt Lake City, Utah, USA
| | - Karen C Schliep
- Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Debs SR, Rothmond DA, Zhu Y, Weickert CS, Purves-Tyson TD. Molecular evidence of altered stress responsivity related to neuroinflammation in the schizophrenia midbrain. J Psychiatr Res 2024; 177:118-128. [PMID: 39004003 DOI: 10.1016/j.jpsychires.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Stress and inflammation are risk factors for schizophrenia. Chronic psychosocial stress is associated with subcortical hyperdopaminergia, a core feature of schizophrenia. Hyperdopaminergia arises from midbrain neurons, leading us to hypothesise that changes in stress response pathways may occur in this region. To identify whether transcriptional changes in glucocorticoid and mineralocorticoid receptors (NR3C1/GR, NR3C2/MR) or other stress signalling molecules (FKBP4, FKBP5) exist in schizophrenia midbrain, we measured gene expression in the human brain (N = 56) using qRT-PCR. We assessed whether alterations in these mRNAs were related to previously identified high/low inflammatory status. We investigated relationships between stress-related transcripts themselves, and between FKBP5 mRNA, dopaminergic, and glial cell transcripts in diagnostic and inflammatory subgroups. Though unchanged by diagnosis, GR mRNA levels were reduced in high inflammatory compared to low inflammatory schizophrenia cases (p = 0.026). We found no effect of diagnosis or inflammation on MR mRNA. FKBP4 mRNA was decreased and FKBP5 mRNA was increased in schizophrenia (p < 0.05). FKBP5 changes occurred in high inflammatory (p < 0.001), whereas FKBP4 changes occurred in low inflammatory schizophrenia cases (p < 0.05). The decrease in mRNA encoding the main stress receptor (GR), as well as increased transcript levels of the stress-responsive negative regulator (FKBP5), may combine to blunt the midbrain response to stress in schizophrenia when neuroinflammation is present. Negative correlations between FKBP5 mRNA and dopaminergic transcripts in the low inflammatory subgroup suggest higher levels of FKBP5 mRNA may also attenuate dopaminergic neurotransmission in schizophrenia even when inflammation is absent. We report alterations in GR-mediated stress signalling in the midbrain in schizophrenia.
Collapse
Affiliation(s)
- Sophie R Debs
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Debora A Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Yunting Zhu
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Tertia D Purves-Tyson
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia.
| |
Collapse
|
11
|
Zhang H, Zhang Y, Xu K, Wang L, Zhou X, Yang M, Xie J, Li H. Inhibition of FLT1 Attenuates Neurodevelopmental Abnormalities and Cognitive Impairment in Offspring Caused by Maternal Prenatal Stress. Appl Biochem Biotechnol 2024; 196:4900-4913. [PMID: 37979086 DOI: 10.1007/s12010-023-04774-6] [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] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
Fms-like tyrosine kinase 1 (FLT1) has been shown to regulate processes such as angiogenesis, neurogenesis, and cognitive impairment. However, the role of FLT1 in prenatal stress (PS) is unclear. The purpose of this study was to investigate the role of FLT1 in PS mothers and their offspring. Wire mesh restrainers were used to construct PS rat model. The levels of FLT1, IL-1β, IL-6, and ROS in clinical samples and rat samples were detected by qRT-PCR, ELisa kit, and DCFH-DA fluorescence kit. Morris water maze assay and forced swimming assay were used to test the cognitive function of offspring young rats. The apoptosis level of hippocampal neurons and the expression of NMDARs were detected by MTT assay, TUNEL assay, and Western blot. The results showed that FLT1 was upregulated in PS mothers and positively correlated with PS degree. The level of FLT1 was elevated in PS model rats. Knockdown of FLT1 reduced maternal ROS and MDA levels and increased SOD levels in PS rats. Knockdown of FLT1 also reduced the secretion of IL-1β, IL-6, and cortisol in PS rats. Inhibition of FTL1 alleviated cognitive impairment in PS offspring pups. Inhibition of FTL1 reduced hippocampal neuronal apoptosis and increased the expression of NMDARs in PS progeny. In conclusions, we demonstrated that knockdown of FLT1 inhibits maternal oxidative stress, inflammation, and cortisol secretion in PS rats. In addition, knockdown of FLT1 also alleviated cognitive dysfunction and neurodevelopmental abnormalities in PS offspring pups.
Collapse
Affiliation(s)
- Huifang Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Yudan Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Kaixuan Xu
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Lawen Wang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Xin Zhou
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Mingge Yang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Jiangli Xie
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Hui Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China.
- Division of Neonatology, The Affiliated Children Hospital of Xi'an Jiaotong University, Shaanxi, 710003, China.
| |
Collapse
|
12
|
Pike MR, Lipner E, O'Brien KJ, Breen EC, Cohn BA, Cirillo PM, Krigbaum NY, Kring AM, Olino TM, Alloy LB, Ellman LM. Prenatal maternal Inflammation, childhood cognition and adolescent depressive symptoms. Brain Behav Immun 2024; 119:908-918. [PMID: 38761818 PMCID: PMC11844254 DOI: 10.1016/j.bbi.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 04/10/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND Accumulating evidence indicates that higher prenatal maternal inflammation is associated with increased depression risk in adolescent and adult-aged offspring. Prenatal maternal inflammation (PNMI) may increase the likelihood for offspring to have lower cognitive performance, which, in turn, may heighten risk for depression onset. Therefore, this study explored the potential mediating role of childhood cognitive performance in the relationship between PNMI and adolescent depressive symptoms in offspring. METHODS Participants included 696 mother-offspring dyads from the Child Health and Development Studies (CHDS) cohort. Biomarkers of maternal inflammation [interleukin (IL)-6, IL-8, IL-1 receptor antagonist (IL-1RA) and soluble TNF receptor-II (sTNF-RII)] were assayed from first (T1) and second trimester (T2) sera. Childhood (ages 9-11) cognitive performance was assessed via standardized Peabody Picture Vocabulary Test (PPVT), a measure of receptive vocabulary correlated with general intelligence. Adolescent (ages 15-17) depressive symptoms were assessed via self-report. RESULTS There were no significant associations between T1 biomarkers and childhood PPVT or adolescent depressive symptoms. Higher T2 IL1-RA was directly associated with lower childhood PPVT (b = -0.21, SE = 0.08, t = -2.55, p = 0.01), but not with adolescent depressive symptoms. T2 IL-6 was not directly associated with childhood PPVT, but higher T2 IL-6 was directly associated at borderline significance with greater depressive symptoms in adolescence (b = 0.05, SE = 0.03, t = 1.96, p = 0.05). Lower childhood PPVT predicted significantly higher adolescent depressive symptoms (b = -0.07, SE = 0.02, t = -2.99, p < 0.01). There was a significant indirect effect of T2 IL-1RA on adolescent depressive symptoms via childhood PPVT (b = 0.03, 95 % CI = 0.002-0.03) indicating a partially mediated effect. No significant associations were found with T2 sTNF-RII nor IL-8. CONCLUSIONS Lower childhood cognitive performance, such as that indicated by a lower PPVT score, represents a potential mechanism through which prenatal maternal inflammation contributes to adolescent depression risk in offspring.
Collapse
Affiliation(s)
- Madeline R Pike
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA.
| | - Emily Lipner
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Kathleen J O'Brien
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Elizabeth C Breen
- Cousins Center for Psychoneuroimmunology, University of California-Los Angeles, 300 Medical Plaza, Suite 3306, Los Angeles, CA 90095-7076, USA
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Ann M Kring
- University of California, Berkeley, Department of Psychology, 2121 Berkeley Way, Berkeley, CA 94720, USA
| | - Thomas M Olino
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Lauren B Alloy
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Lauren M Ellman
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| |
Collapse
|
13
|
Lipner E, Mac Giollabhui N, Breen EC, Cohn BA, Krigbaum NY, Cirillo PM, Olino TM, Alloy LB, Ellman LM. Sex-Specific Pathways From Prenatal Maternal Inflammation to Adolescent Depressive Symptoms. JAMA Psychiatry 2024; 81:498-505. [PMID: 38324324 PMCID: PMC10851141 DOI: 10.1001/jamapsychiatry.2023.5458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 02/08/2024]
Abstract
Importance Prenatal maternal inflammation has been associated with major depressive disorder in offspring in adulthood as well as with internalizing and externalizing symptoms in childhood; however, the association between prenatal inflammation and offspring depression in adolescence has yet to be examined. Objective To determine whether maternal levels of inflammatory biomarkers during pregnancy are associated with depressive symptomatology in adolescent-aged offspring and to examine how gestational timing, offspring sex, and childhood psychiatric symptoms impact these associations. Design, Setting, and Participants This was an observational study of a population-based birth cohort from the Child Health and Development Studies (CHDS), which recruited almost all mothers receiving obstetric care from the Kaiser Foundation Health Plan (KFHP) in Alameda County, California, between June 1959 and September 1966. Pregnancy data and blood sera were collected from mothers, and offspring psychiatric symptom data were collected in childhood (ages 9-11 years) and adolescence (ages 15-17 years). Mother-offspring dyads with available maternal prenatal inflammatory biomarkers during first and/or second trimesters and offspring depressive symptom data at adolescent follow-up were included. Data analyses took place between March 2020 and June 2023. Exposures Levels of inflammatory biomarkers (interleukin 6 [IL-6], IL-8, IL-1 receptor antagonist [IL-1RA], and soluble tumor necrosis factor receptor-II) assayed from maternal sera in the first and second trimesters of pregnancy. Main Outcomes and Measures Self-reported depressive symptoms at adolescent follow-up. Results A total of 674 mothers (mean [SD] age, 28.1 [5.9] years) and their offspring (350 male and 325 female) were included in this study. Higher second trimester IL-6 was significantly associated with greater depressive symptoms in offspring during adolescence (b, 0.57; SE, 0.26); P = .03). Moderated mediation analyses showed that childhood externalizing symptoms significantly mediated the association between first trimester IL-6 and adolescent depressive symptoms in male offspring (b, 0.18; 95% CI, 0.02-0.47), while childhood internalizing symptoms mediated the association between second trimester IL-1RA and adolescent depressive symptoms in female offspring (b, 0.80; 95% CI, 0.19-1.75). Conclusions and Relevance In this study, prenatal maternal inflammation was associated with depressive symptoms in adolescent-aged offspring. The findings of the study suggest that pathways to adolescent depressive symptomatology from prenatal risk factors may differ based on both the timing of exposure to prenatal inflammation and offspring sex.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Naoise Mac Giollabhui
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
- Department of Psychiatry, Massachusetts General Hospital, Boston
| | - Elizabeth C. Breen
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles
| | - Barbara A. Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Nickilou Y. Krigbaum
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Piera M. Cirillo
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Thomas M. Olino
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Lauren M. Ellman
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Chen RJ, Nabila A, Gal Toth J, Stuhlmann H, Toth M. The chemokine XCL1 functions as a pregnancy hormone to program offspring innate anxiety. Brain Behav Immun 2024; 118:178-189. [PMID: 38428650 PMCID: PMC11044916 DOI: 10.1016/j.bbi.2024.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Elevated levels of cytokines in maternal circulation increase the offspring's risk for neuropsychiatric disease. Because of their low homeostatic levels, circulating maternal cytokines during normal pregnancies have not been considered to play a role in fetal brain development and offspring behavior. Here we report that the T/NK cell chemotactic cytokine XCL1, a local paracrine immune signal, can function as a pregnancy hormone and is required for the proper development of placenta and male offspring approach-avoidance behavior. We found that circulating XCL1 levels were at a low pregestational level throughout pregnancy except for a midgestational rise and fall. Blunted elevation in maternal plasma XCL1 in dams with a genetic 5HT1A receptor deficit or following neutralization by anti-XCL1 antibodies increased the expression of tissue damage associated factors in WT fetal placenta and led to increased innate anxiety and stress reactivity in the WT male offspring. Therefore, chemokines like XCL1 may act as pregnancy hormones to regulate placenta development and offspring emotional behavior.
Collapse
Affiliation(s)
- Rosa J Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anika Nabila
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Judit Gal Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi Stuhlmann
- Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miklos Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
15
|
Abrishamcar S, Zhuang B, Thomas M, Gladish N, MacIsaac J, Jones M, Simons E, Moraes T, Mandhane P, Brook J, Subbarao P, Turvey S, Chen E, Miller G, Kobor M, Huels A. Association between Maternal Perinatal Stress and Depression on Infant DNA Methylation in the First Year of Life. RESEARCH SQUARE 2024:rs.3.rs-3962429. [PMID: 38562779 PMCID: PMC10984027 DOI: 10.21203/rs.3.rs-3962429/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Maternal stress and depression during pregnancy and the first year of the infant's life affect a large percentage of mothers. Maternal stress and depression have been associated with adverse fetal and childhood outcomes as well as differential child DNA methylation (DNAm). However, the biological mechanisms connecting maternal stress and depression to poor health outcomes in children are still largely unknown. Here we aim to determine whether prenatal stress and depression are associated with changes in cord blood mononuclear cell DNAm (CBMC-DNAm) in newborns (n = 119) and whether postnatal stress and depression are associated with changes in peripheral blood mononuclear cell DNAm (PBMC-DNAm) in children of 12 months of age (n = 113) from the Canadian Healthy Infant Longitudinal Development (CHILD) cohort. Stress was measured using the 10-item Perceived Stress Scale (PSS) and depression was measured using the Center for Epidemiologic Studies Depression Questionnaire (CESD). Both stress and depression were measured at 18 weeks and 36 weeks of pregnancy and six months and 12 months postpartum. We conducted epigenome-wide association studies (EWAS) using robust linear regression followed by a sensitivity analysis in which we bias-adjusted for inflation and unmeasured confounding using the bacon and cate methods. To investigate the cumulative effect of maternal stress and depression, we created composite prenatal and postnatal adversity scores. We identified a significant association between prenatal stress and differential CBMC-DNAm at 8 CpG sites and between prenatal depression and differential CBMC-DNAm at 2 CpG sites. Additionally, we identified a significant association between postnatal stress and differential PBMC-DNAm at 8 CpG sites and between postnatal depression and differential PBMC-DNAm at 11 CpG sites. Using our composite scores, we further identified 2 CpG sites significantly associated with prenatal adversity and 7 CpG sites significantly associated with postnatal adversity. Several of the associated genes, including PLAGL1, HYMAI, BRD2, and ERC2 have been implicated in adverse fetal outcomes and neuropsychiatric disorders. This suggested that differential DNAm may play a role in the relationship between maternal mental health and child health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Anke Huels
- Rollins School of Public Health, Emory University
| |
Collapse
|
16
|
Nishigori H, Nishigori T, Suzuki T, Mori M, Yamada M, Isogami H, Murata T, Kyozuka H, Ogata Y, Sato A, Metoki H, Shinoki K, Yasumura S, Hosoya M, Hashimoto K, Fujimori K. Maternal prenatal and postnatal psychological distress trajectories and impact on cognitive development in 4-year-old children: the Japan Environment and Children's Study. J Dev Orig Health Dis 2023; 14:781-794. [PMID: 38327072 DOI: 10.1017/s2040174424000011] [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] [Indexed: 02/09/2024]
Abstract
Maternal prenatal and postnatal psychological distress, including depression and anxiety, may affect children's cognitive development. However, the findings have been inconsistent. We aimed to use the dataset from the Japan Environment and Children's Study, a nationwide prospective birth cohort study, to examine this association. We evaluated the relationship between the maternal six-item version of the Kessler Psychological Distress Scale (K6) scores and cognitive development among children aged 4 years. K6 was administered twice during pregnancy (M-T1; first half of pregnancy, M-T2; second half of pregnancy) and 1 year postpartum (C-1y). Cognitive development was assessed by trained testers, using the Kyoto Scale of Psychological Development 2001. Multiple regression analysis was performed with the group with a K6 score ≤ 4 for both M-T1 and M-T2 and C-1y as a reference. Records from 1,630 boys and 1,657 girls were analyzed. In the group with K6 scores ≥ 5 in both M-T1 and M-T2 and C-1Y groups, boys had significantly lower developmental quotients (DQ) in the language-social developmental (L-S) area (partial regression coefficient: -4.09, 95% confidence interval: -6.88 - -1.31), while girls did not differ significantly in DQ for the L-S area. Among boys and girls, those with K6 scores ≤ 4 at any one or two periods during M-T1, M-T2, or C-1y did not have significantly lower DQ for the L-S area. Persistent maternal psychological distress from the first half of pregnancy to 1 year postpartum had a disadvantageous association with verbal cognitive development in boys, but not in girls aged 4 years.
Collapse
Affiliation(s)
- Hidekazu Nishigori
- Department of Development and Environmental Medicine, Fukushima Medical Center for Children and Women, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
| | - Toshie Nishigori
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Pediatrics, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Taeko Suzuki
- Department of Development and Environmental Medicine, Fukushima Medical Center for Children and Women, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Midwifery and Maternal Nursing, Fukushima Medical University School of Nursing, Fukushima, Japan
| | - Miyuki Mori
- Department of Development and Environmental Medicine, Fukushima Medical Center for Children and Women, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Midwifery and Maternal Nursing, Fukushima Medical University School of Nursing, Fukushima, Japan
| | - Mika Yamada
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Pediatrics, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hirotaka Isogami
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tsuyoshi Murata
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hyo Kyozuka
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuka Ogata
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
| | - Akiko Sato
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kosei Shinoki
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
| | - Seiji Yasumura
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Public Health, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mitsuaki Hosoya
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Pediatrics, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Koichi Hashimoto
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Pediatrics, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Keiya Fujimori
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima, Japan
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
17
|
Martín-Cuevas C, Ramos-Herrero VD, Crespo-Facorro B, Sánchez-Hidalgo AC. Prenatal risk factors and postnatal cannabis exposure: Assessing dual models of schizophrenia-like rodents. Neurosci Biobehav Rev 2023; 154:105409. [PMID: 37783300 DOI: 10.1016/j.neubiorev.2023.105409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/04/2023] [Accepted: 09/24/2023] [Indexed: 10/04/2023]
Abstract
Schizophrenia (SCZ) is a multifactorial neurodevelopmental disorder caused by genetic and environmental alterations, especially during prenatal stages. On the other hand, cannabis consumption in adolescence has been also linked to an increased risk of developing SCZ. The combination of both hits has been proposed as the dual hit hypothesis of SCZ. We systematically reviewed prenatal environmental alterations and cannabis consumption during adolescence that are associated with an increased risk of SCZ, following the PRISMA model. The analysis focused on dual animal models where the first hit is prenatal environmental exposure and the second hit consists of postnatal cannabis exposure. The articles were evaluated by three independent reviewers based on inclusion criteria. We extracted the first author´s name, year, model species, sex and analysis. The articles reported on dual murine models and their effects on weight, behavior, genetics, electrophysiology and brain structure and function. We conclude that the defects caused by the dual hits depend on the sex of the model, as well as type of hits.
Collapse
Affiliation(s)
- Celia Martín-Cuevas
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, 41013 Seville, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain.
| | - Víctor Darío Ramos-Herrero
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, 41013 Seville, Spain.
| | - Benedicto Crespo-Facorro
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, 41013 Seville, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain; Department of Psychiatry, School of Medicine, University of Sevilla, Manuel Siurot AV, 41013 Seville, Spain.
| | - Ana C Sánchez-Hidalgo
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, 41013 Seville, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
18
|
Abramova O, Morozova A, Zubkov E, Ushakova V, Zorkina Y, Proshin AT, Storozheva Z, Gurina O, Chekhonin V. Ultrasound-Induced Prenatal Stress: New Possibilities for Modeling Mental Disorders. Dev Neurosci 2023; 46:237-261. [PMID: 37857257 PMCID: PMC11251674 DOI: 10.1159/000534687] [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: 03/01/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
The development of animal models of mental disorders is an important task since such models are useful for studying the neurobiological mechanisms of psychopathologies and for trial of new therapeutic drugs. One way to model pathologies of the nervous system is to impair fetal neurodevelopment through stress of the pregnant future mother, or prenatal stress (PS). The use of variable frequency ultrasound (US) in rodents is a promising method of imitating psychological stress, to which women in modern society are most often subjected. The aim of our study was to investigate the effect of PS induced by exposure to variable frequency ultrasound (US PS) throughout the gestational period on the adult rat offspring, namely, to identify features of behavioral alterations and neurochemical brain parameters that can be associated with certain mental disorders in humans, to determine the possibility of creating a new model of psychopathology. Our study included a study of some behavioral characteristics of male and female rats in the elevated plus maze, open-field test, object recognition test, social interaction test, sucrose preference test, latent inhibition test, Morris water maze, forced swimming test, acoustic startle reflex, and prepulse inhibition tests. We also determined the activity of the serotonergic, dopaminergic, and noradrenergic neurotransmitter systems in the hippocampus and frontal cortex by HPLC-ED. Concentration of norepinephrine, dopamine, DOPAC, serotonin, and HIAA, as well as DOPAC/dopamine and HIAA/serotonin ratios were determined. A correlation analysis of behavioral and neurochemical parameters in male and female rats was performed based on the data obtained. The results of the study showed that US PS altered the behavioral phenotype of the rat offspring. US PS increased the level of anxious behavior, impaired orientation-research behavior, increased grooming activity, decreased the desire for social contacts, shifted behavioral reactions from social interaction to interaction with inanimate objects, impaired latent inhibition, and decreased the startle reflex. US PS activated the serotonergic, dopaminergic, and noradrenergic neurotransmitter systems of the rat frontal cortex and hippocampus. A correlation between neurochemical and behavioral parameters was revealed. Our study showed that US PS leads to a certain dysfunction on behavioral and neurochemical levels in rats that is most closely associated with symptoms of schizophrenia or autism. We hypothesize that this could potentially be an indicator of face validity for a model of psychopathology based on neurodevelopmental impairment.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
- Mental-health Clinic No. 1 named after N.A. Alekseev, Moscow, Russia
| | - Anna Morozova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
- Mental-health Clinic No. 1 named after N.A. Alekseev, Moscow, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Valeria Ushakova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
- Mental-health Clinic No. 1 named after N.A. Alekseev, Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
- Mental-health Clinic No. 1 named after N.A. Alekseev, Moscow, Russia
| | - Andrey T. Proshin
- Laboratory of General Physiology of Functional Systems, P.K. Anokhin Institute of Normal Physiology, Moscow, Russia
| | - Zinaida Storozheva
- Laboratory of Functional Neurochemistry, P.K. Anokhin Institute of Normal Physiology, Moscow, Russia
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
19
|
Baines KJ, West RC. Sex differences in innate and adaptive immunity impact fetal, placental, and maternal health†. Biol Reprod 2023; 109:256-270. [PMID: 37418168 DOI: 10.1093/biolre/ioad072] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023] Open
Abstract
The differences between males and females begin shortly after birth, continue throughout prenatal development, and eventually extend into childhood and adult life. Male embryos and fetuses prioritize proliferation and growth, often at the expense of the fetoplacental energy reserves. This singular focus on growth over adaptability leaves male fetuses and neonates vulnerable to adverse outcomes during pregnancy and birth and can have lasting impacts throughout life. Beyond this prioritization of growth, male placentas and fetuses also respond to infection and inflammation differently than female counterparts. Pregnancies carrying female fetuses have a more regulatory immune response, whereas pregnancies carrying male fetuses have a stronger inflammatory response. These differences can be seen as early as the innate immune response with differences in cytokine and chemokine signaling. The sexual dimorphism in immunity then continues into the adaptive immune response with differences in T-cell biology and antibody production and transfer. As it appears that these sex-specific differences are amplified in pathologic pregnancies, it stands to reason that differences in the placental, fetal, and maternal immune responses in pregnancy contribute to increased male perinatal morbidity and mortality. In this review, we will describe the genetic and hormonal contributions to the sexual dimorphism of fetal and placental immunity. We will also discuss current research efforts to describe the sex-specific differences of the maternal-fetal interface and how it impacts fetal and maternal health.
Collapse
Affiliation(s)
- Kelly J Baines
- Anatomy, Physiology, Pharmacology Department, Auburn University, Auburn, AL 36849, USA
| | - Rachel C West
- Anatomy, Physiology, Pharmacology Department, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
20
|
Marszalek-Grabska M, Gawel K, Kosheva N, Kocki T, Turski WA. Developmental Exposure to Kynurenine Affects Zebrafish and Rat Behavior. Cells 2023; 12:2224. [PMID: 37759447 PMCID: PMC10526278 DOI: 10.3390/cells12182224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Proper nutrition and supplementation during pregnancy and breastfeeding are crucial for the development of offspring. Kynurenine (KYN) is the central metabolite of the kynurenine pathway and a direct precursor of other metabolites that possess immunoprotective or neuroactive properties, with the ultimate effect on fetal neurodevelopment. To date, no studies have evaluated the effects of KYN on early embryonic development. Thus, the aim of our study was to determine the effect of incubation of larvae with KYN in different developmental periods on the behavior of 5-day-old zebrafish. Additionally, the effects exerted by KYN administered on embryonic days 1-7 (ED 1-7) on the behavior of adult offspring of rats were elucidated. Our study revealed that the incubation with KYN induced changes in zebrafish behavior, especially when zebrafish embryos or larvae were incubated with KYN from 1 to 72 h post-fertilization (hpf) and from 49 to 72 hpf. KYN administered early during pregnancy induced subtle differences in the neurobehavioral development of adult offspring. Further research is required to understand the mechanism of these changes. The larval zebrafish model can be useful for studying disturbances in early brain development processes and their late behavioral consequences. The zebrafish-medium system may be applicable in monitoring drug metabolism in zebrafish.
Collapse
Affiliation(s)
- Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University, Jaczewskiego 8b, 20-090 Lublin, Poland; (K.G.); (N.K.); (T.K.); (W.A.T.)
| | | | | | | | | |
Collapse
|
21
|
Mawson ER, Morris BJ. A consideration of the increased risk of schizophrenia due to prenatal maternal stress, and the possible role of microglia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110773. [PMID: 37116354 DOI: 10.1016/j.pnpbp.2023.110773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Schizophrenia is caused by interaction of a combination of genetic and environmental factors. Of the latter, prenatal exposure to maternal stress is reportedly associated with elevated disease risk. The main orchestrators of inflammatory processes within the brain are microglia, and aberrant microglial activation/function has been proposed to contribute to the aetiology of schizophrenia. Here, we evaluate the epidemiological and preclinical evidence connecting prenatal stress to schizophrenia risk, and consider the possible mediating role of microglia in the prenatal stress-schizophrenia relationship. Epidemiological findings are rather consistent in supporting the association, albeit they are mitigated by effects of sex and gestational timing, while the evidence for microglial activation is more variable. Rodent models of prenatal stress generally report lasting effects on offspring neurobiology. However, many uncertainties remain as to the mechanisms underlying the influence of maternal stress on the developing foetal brain. Future studies should aim to characterise the exact processes mediating this aspect of schizophrenia risk, as well as focussing on how prenatal stress may interact with other risk factors.
Collapse
Affiliation(s)
- Eleanor R Mawson
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
22
|
Environmental Risk Factors and Cognitive Outcomes in Psychosis: Pre-, Perinatal, and Early Life Adversity. Curr Top Behav Neurosci 2023; 63:205-240. [PMID: 35915384 PMCID: PMC9892366 DOI: 10.1007/7854_2022_378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Risk for psychosis begins to accumulate as early as the fetal period through exposure to obstetric complications like fetal hypoxia, maternal stress, and prenatal infection. Stressors in the postnatal period, such as childhood trauma, peer victimization, and neighborhood-level adversity, further increase susceptibility for psychosis. Cognitive difficulties are among the first symptoms to emerge in individuals who go on to develop a psychotic disorder. We review the relationship between pre-, perinatal, and early childhood adversities and cognitive outcomes in individuals with psychosis. Current evidence shows that the aforementioned environmental risk factors may be linked to lower overall intelligence and executive dysfunction, beginning in the premorbid period and persisting into adulthood in individuals with psychosis. It is likely that early life stress contributes to cognitive difficulties in psychosis through dysregulation of the body's response to stress, causing changes such as increased cortisol levels and chronic immune activation, which can negatively impact neurodevelopment. Intersectional aspects of identity (e.g., sex/gender, race/ethnicity), as well as gene-environment interactions, likely inform the developmental cascade to cognitive difficulties throughout the course of psychotic disorders and are reviewed below. Prospective studies of birth cohorts will serve to further clarify the relationship between early-life environmental risk factors and cognitive outcomes in the developmental course of psychotic disorders. Specific methodological recommendations are provided for future research.
Collapse
|
23
|
Zhang YS, Rao WW, Zhang LL, Jia HX, Bi H, Wang HL, Balbuena L, Li KQ, Xiang YT. Incidence rate of schizophrenia after the Tangshan earthquake in China: a 44-year retrospective birth cohort study. Transl Psychiatry 2022; 12:365. [PMID: 36068195 PMCID: PMC9448782 DOI: 10.1038/s41398-022-02125-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022] Open
Abstract
Preliminary evidence indicates that natural disasters are associated with an increased risk for schizophrenia. With few longitudinal studies on earthquakes, this retrospective cohort study examined exposure to the 1976 Tangshan earthquake and the subsequent risk of schizophrenia. Population counts and visits to all nine psychiatric hospitals in Tangshan city were collected. We created three cohort groups by earthquake exposure: infant (August 1972 to July 1976 births), fetal (August 1976 to May 1977 births), and unexposed (June 1977 to May 1981 births). The cumulative incidence of schizophrenia in each cohort was calculated by dividing the number of schizophrenia patients by total births in the corresponding period. Altogether, 6424 schizophrenia patients were identified, with 2786 in the infant group, 663 in the fetal group, and 2975 in the unexposed group. The crude cumulative incidence of schizophrenia in the infant, fetal and unexposed groups were 7.64 (95% confidence interval [CI] = 7.36-7.92), 9.07 (95% CI = 8.38-9.76), and 7.40 (95% CI = 7.13-7.66) per thousand population respectively. Adjusted for mortality, the corresponding figures were 7.73 (95% CI = 7.44-8.01), 9.30 (95% CI = 8.60-10.01) and 7.44 (95% CI = 7.18-7.71) per thousand population respectively. The mortality-adjusted risk ratio (aRR) was 1.25 (95% CI = 1.15-1.36) between fetal and unexposed groups (χ2 = 27.31, P < 0.001). Males exposed as infants did not differ from the unexposed in cumulative schizophrenia incidence. People with fetal exposure to the 1976 earthquake had 25% higher risk of developing schizophrenia compared to unexposed counterparts.
Collapse
Affiliation(s)
- Yun-Shu Zhang
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Hebei Provincial Mental Health Center, Baoding, Hebei Province, China
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
| | - Wen-Wang Rao
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
| | - Li-Li Zhang
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Hebei Provincial Mental Health Center, Baoding, Hebei Province, China
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
| | - Hong-Xue Jia
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
- College of Public Health, Hebei University, Baoding, Hebei province, China
| | - Hao Bi
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Hebei Provincial Mental Health Center, Baoding, Hebei Province, China
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
| | - Hai-Long Wang
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Hebei Provincial Mental Health Center, Baoding, Hebei Province, China
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China
| | - Lloyd Balbuena
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ke-Qing Li
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Hebei Provincial Mental Health Center, Baoding, Hebei Province, China.
- Institute of Mental Health, Hebei Provincial Mental Health Center, Baoding, Hebei province, China.
| | - Yu-Tao Xiang
- Unit of Psychiatry, Department of Public Health and Medicinal Administration, & Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China.
- Centre for Cognitive and Brain Sciences, University of Macau, Macao SAR, China.
- Institute of Advanced Studies in Humanities and Social Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
24
|
Lipner E, Murphy SK, Breen EC, Cohn BA, Krigbaum NY, Cirillo PM, Alloy LB, Ellman LM. Infection and higher cortisol during pregnancy and risk for depressive symptoms in adolescent offspring. Psychoneuroendocrinology 2022; 141:105755. [PMID: 35429699 PMCID: PMC9149123 DOI: 10.1016/j.psyneuen.2022.105755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 02/24/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Prenatal infection, particularly at mid-gestation, has been associated with various psychopathological outcomes in offspring; however, findings linking prenatal infection to offspring depression outcomes have been mixed. Previous research indicates that it may be the co-occurrence of prenatal adversities (e.g., infection and stress) that are associated with depression outcomes in offspring. Nevertheless, no study to date has investigated whether higher levels of biomarkers linked to prenatal stress (e.g., cortisol) in the presence of infection may account for these outcomes. Participants were drawn from the Child Health and Development Studies (CHDS), a prospective, longitudinal study of pregnant women and their offspring. The present study included mother-offspring dyads from the Adolescent Study, a subsample of the CHDS cohort, whose offspring were assessed in adolescence and whose mothers also provided sera to be assayed for cortisol (n = 695). Hierarchical multivariable regressions were conducted to examine whether maternal cortisol during the first and second trimesters of pregnancy interacted with maternal infection to predict increased risk for symptoms of depression in adolescent offspring. There was a significant interaction of second trimester infection and higher cortisol on offspring depression scores during adolescence, controlling for maternal education (p = 0.04). Findings suggest that higher maternal cortisol may sensitize mothers and their offspring to the disruptive influences of infection during mid-pregnancy, conferring greater risk of depressive symptomatology in offspring.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Shannon K Murphy
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Elizabeth C Breen
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, University of California, 300 Medical Plaza, Los Angeles, CA 90095, USA
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Ste. B, Berkeley, CA 94709
| | - Lauren B Alloy
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | - Lauren M Ellman
- Department of Psychology and Neuroscience, Temple University, Weiss Hall, 1701 N. 13th Street, Philadelphia, PA 19122, USA.
| |
Collapse
|
25
|
DeRosa H, Richter T, Wilkinson C, Hunter RG. Bridging the Gap Between Environmental Adversity and Neuropsychiatric Disorders: The Role of Transposable Elements. Front Genet 2022; 13:813510. [PMID: 35711940 PMCID: PMC9196244 DOI: 10.3389/fgene.2022.813510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/13/2022] [Indexed: 12/21/2022] Open
Abstract
Long regarded as “junk DNA,” transposable elements (TEs) have recently garnered much attention for their role in promoting genetic diversity and plasticity. While many processes involved in mammalian development require TE activity, deleterious TE insertions are a hallmark of several psychiatric disorders. Moreover, stressful events including exposure to gestational infection and trauma, are major risk factors for developing psychiatric illnesses. Here, we will provide evidence demonstrating the intersection of stressful events, atypical TE expression, and their epigenetic regulation, which may explain how neuropsychiatric phenotypes manifest. In this way, TEs may be the “bridge” between environmental perturbations and psychopathology.
Collapse
Affiliation(s)
- Holly DeRosa
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Troy Richter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Cooper Wilkinson
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Richard G Hunter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
26
|
Bernard NK, Bogat GA, Kashy DA, Lonstein JS, Levendosky AA. Prenatal and postnatal intimate partner violence, depression, and infant-mother touch. Infant Behav Dev 2022; 67:101703. [DOI: 10.1016/j.infbeh.2022.101703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022]
|
27
|
Impacts of a perinatal exposure to manganese coupled with maternal stress in rats: Tests of untrained behaviors. Neurotoxicol Teratol 2022; 91:107088. [PMID: 35278630 PMCID: PMC9133146 DOI: 10.1016/j.ntt.2022.107088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/01/2022] [Accepted: 03/06/2022] [Indexed: 11/21/2022]
Abstract
Manganese (Mn), an element that naturally occurs in the environment, has been shown to produce neurotoxic effects on the developing young when levels exceed physiological requirements. To evaluate the effects of this chemical in combination with non-chemical factors pregnant Long-Evans rats were treated with 0, 2, or 4 mg/mL Mn in their drinking water from gestational day (GD) 7 to postnatal day (PND) 22. Half of the dams received a variable stress protocol from GD13 to PND9, that included restraint, small cage with reduced bedding, exposure to predator odor, intermittent intervals of white noise, lights on for 24 h, intermittent intervals of lights on during dark cycle and cages with grid floors and reduced bedding. One male and one female offspring from each litter were tested to assess untrained behavior. Ultrasonic vocalizations (USV) were recorded from PND13 pups while they were isolated from the litter. Locomotor activity (MA) was measured in figure-eight mazes at PND 17, 29, and 79 (different set of rats at each time point). Social approach (SA) was tested at PND48. Acoustic startle response (ASR) and pre-pulse inhibition (PPI) were measured starting at PND58. At PND53 a sweetness preference for a chocolate flavored milk solution was assessed. There were sex related differences on several parameters for the USVs. There was also a Mn by stress by sex interaction with the females from the 4 mg/mL stressed dams having more frequency modulated (FM) call elements than the 4 mg/mL non-stressed group. There was an effect of Mn on motor activity but only at PND29 with the 2 mg/mL group having higher counts than the 0 mg/mL group. The social approach test showed sex differences for both the habituation and test phase. There was an effect of Mn, with the 4 mg/mL males having a greater preference for the stimulus rat than did the 0 mg/mL males. There was also a stress by sex interaction. The ASR and PPI had only a sex effect. Thus, with only the FM call elements having a Mn by stress effect, and the PND29 MA and SA preference index having a Mn effect but at different doses requires further investigation.
Collapse
|
28
|
Momen NC, Robakis T, Liu X, Reichenberg A, Bergink V, Munk-Olsen T. In utero exposure to antipsychotic medication and psychiatric outcomes in the offspring. Neuropsychopharmacology 2022; 47:759-766. [PMID: 34750566 PMCID: PMC8782838 DOI: 10.1038/s41386-021-01223-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 02/03/2023]
Abstract
Information on neurodevelopmental effects of antenatal exposure to antipsychotics is limited to 10 studies, all examining children up to 5 years of age or less. The paper aimed to investigate the association between in utero exposure to antipsychotics and psychiatric outcomes in children using Danish nationwide registers. In total, 9011 liveborn singletons born 1998-2015 in Denmark whose mothers took antipsychotic medication before pregnancy were identified. Children whose mothers continued to take antipsychotics during pregnancy were compared with children of mothers who discontinued antipsychotics before pregnancy. As a negative control, paternal antipsychotic use in the same window was investigated. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using Cox proportional hazards regression for the primary outcome of psychiatric disorders, as well for subcategories of psychiatric disorders. In total, 9.9% of children in the discontinuation group and 11.0% of children in the continuation group received a psychiatric disorder diagnosis during follow-up. The adjusted HR for psychiatric disorders among offspring in the continuation group compared to the discontinuation group was 1.10 (95% CI 0.93-1.30). For antipsychotic use in the fathers, the HR was 1.05 (95% CI 0.89-1.24). The study does not provide evidence of increased risk of psychiatric disorders among children of women who continue antipsychotic treatment during pregnancy. This was observed after accounting for the underlying risk conferred by maternal psychiatric disorders. This suggests women who need to continue antipsychotic medications during pregnancy can do so without adverse psychiatric outcomes for offspring.
Collapse
Affiliation(s)
- Natalie C. Momen
- grid.7048.b0000 0001 1956 2722National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Thalia Robakis
- grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY USA
| | - Xiaoqin Liu
- grid.7048.b0000 0001 1956 2722National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Abraham Reichenberg
- grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY USA ,grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Seaver Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Veerle Bergink
- grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY USA ,grid.5645.2000000040459992XDepartment of Psychiatry, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Trine Munk-Olsen
- grid.7048.b0000 0001 1956 2722National Centre for Register-based Research, Aarhus University, Aarhus, Denmark ,grid.452548.a0000 0000 9817 5300iPSYCH-Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark ,grid.7048.b0000 0001 1956 2722CIRRAU-Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| |
Collapse
|
29
|
Seeman MV, González-Rodríguez A. Stratification by Sex and Hormone Level When Contrasting Men and Women in Schizophrenia Trials Will Improve Personalized Treatment. J Pers Med 2021; 11:929. [PMID: 34575706 PMCID: PMC8471344 DOI: 10.3390/jpm11090929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Sex and gender differences have been reported in the prevalence, expression, treatment response, and outcome of schizophrenia, but most reports are based on relatively small samples that have not been stratified for the impact of sex hormone levels. This literature review aims to show how women's hormone levels can impact the results of male/female comparisons. METHODS This is a narrative review of data from publications of the last decade. RESULTS Epidemiologic evidence, reports of the impact of hormones on cognition, results of sexually dimorphic responses to treatment, and male/female trajectories of illness over time all suggest that female hormone fluctuations exert major effects on male/female differences in schizophrenia. CONCLUSIONS Information on hormonal status in women participants is rarely available in clinical studies in schizophrenia, which makes male/female comparisons largely uninterpretable. These are the current challenges. Opportunities for individualized treatment are growing, however, and will undoubtedly result in improved outcomes for both women and men in the future.
Collapse
Affiliation(s)
- Mary V. Seeman
- Department of Psychiatry, University of Toronto, #605 260 Heath St. W., Toronto, ON M5P 3L6, Canada
| | - Alexandre González-Rodríguez
- Department of Mental Health, Mutua Terrassa University Hospital, University of Barcelona, 08221 Terrassa, Barcelona, Spain;
| |
Collapse
|
30
|
Early magnetic resonance imaging biomarkers of schizophrenia spectrum disorders: Toward a fetal imaging perspective. Dev Psychopathol 2021; 33:899-913. [PMID: 32489161 DOI: 10.1017/s0954579420000218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is mounting evidence to implicate the intrauterine environment as the initial pathogenic stage for neuropsychiatric disease. Recent developments in magnetic resonance imaging technology are making a multimodal analysis of the fetal central nervous system a reality, allowing analysis of structural and functional parameters. Exposures to a range of pertinent risk factors whether preconception or in utero can now be indexed using imaging techniques within the fetus' physiological environment. This approach may determine the first "hit" required for diseases that do not become clinically manifest until adulthood, and which only have subtle clinical markers during childhood and adolescence. A robust characterization of a "multi-hit" hypothesis may necessitate a longitudinal birth cohort; within this investigative paradigm, the full range of genetic and environmental risk factors can be assessed for their impact on the early developing brain. This will lay the foundation for the identification of novel biomarkers and the ability to devise methods for early risk stratification and disease prevention. However, these early markers must be followed over time: first, to account for neural plasticity, and second, to assess the effects of postnatal exposures that continue to drive the individual toward disease. We explore these issues using the schizophrenia spectrum disorders as an illustrative paradigm. However, given the potential richness of fetal magnetic resonance imaging, and the likely overlap of biomarkers, these concepts may extend to a range of neuropsychiatric conditions.
Collapse
|
31
|
Anglin DM, Ereshefsky S, Klaunig MJ, Bridgwater MA, Niendam TA, Ellman LM, DeVylder J, Thayer G, Bolden K, Musket CW, Grattan RE, Lincoln SH, Schiffman J, Lipner E, Bachman P, Corcoran CM, Mota NB, van der Ven E. From Womb to Neighborhood: A Racial Analysis of Social Determinants of Psychosis in the United States. Am J Psychiatry 2021; 178:599-610. [PMID: 33934608 PMCID: PMC8655820 DOI: 10.1176/appi.ajp.2020.20071091] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The authors examine U.S.-based evidence that connects characteristics of the social environment with outcomes across the psychosis continuum, from psychotic experiences to schizophrenia. The notion that inequitable social and economic systems of society significantly influence psychosis risk through proxies, such as racial minority and immigrant statuses, has been studied more extensively in European countries. While there are existing international reviews of social determinants of psychosis, none to the authors' knowledge focus on factors in the U.S. context specifically-an omission that leaves domestic treatment development and prevention efforts incomplete and underinformed. In this review, the authors first describe how a legacy of structural racism in the United States has shaped the social gradient, highlighting consequential racial inequities in environmental conditions. The authors offer a hypothesized model linking structural racism with psychosis risk through interwoven intermediary factors based on existing theoretical models and a review of the literature. Neighborhood factors, cumulative trauma and stress, and prenatal and perinatal complications were three key areas selected for review because they reflect social and environmental conditions that may affect psychosis risk through a common pathway shaped by structural racism. The authors describe evidence showing that Black and Latino people in the United States suffer disproportionately from risk factors within these three key areas, in large part as a result of racial discrimination and social disadvantage. This broad focus on individual and community factors is intended to provide a consolidated space to review this growing body of research and to guide continued inquiries into social determinants of psychosis in U.S. contexts.
Collapse
Affiliation(s)
- Deidre M Anglin
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Sabrina Ereshefsky
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Mallory J Klaunig
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Miranda A Bridgwater
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Tara A Niendam
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Lauren M Ellman
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Jordan DeVylder
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Griffin Thayer
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Khalima Bolden
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Christie W Musket
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Rebecca E Grattan
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Sarah Hope Lincoln
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Jason Schiffman
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Emily Lipner
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Peter Bachman
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Cheryl M Corcoran
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Natália B Mota
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| | - Els van der Ven
- Department of Psychology, City College of New York, City University of New York, New York (Anglin, Thayer); Graduate Center, City University of New York (Anglin); Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento (Ereshefsky, Niendam, Bolden, Grattan); Department of Psychology, University of Maryland, Baltimore County (Klaunig, Bridgwater, Schiffman); Department of Psychology, Temple University, Philadelphia (Ellman, Lipner); Graduate School of Social Service, Fordham University, New York (DeVylder); Department of Psychology (Musket) and Department of Psychiatry (Bachman), University of Pittsburgh, Pittsburgh; ISN Innovations, Institute for Social Neuroscience, Ivanhoe, Australia (Grattan); Department of Psychological Sciences, Case Western Reserve University, Cleveland (Lincoln); Department of Psychological Science, University of California, Irvine (Schiffman); Department of Psychiatry, Icahn School of Medicine, New York, and James J. Peters VA Medical Center, Bronx, N.Y. (Corcoran); Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil (Mota); Mailman School of Public Health, Columbia University, New York (van der Ven); School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (van der Ven); Department of Clinical, Neuro, and Developmental Psychology, Vrije Universiteit (VU) Amsterdam, Amsterdam (van der Ven)
| |
Collapse
|
32
|
Developmental stress has sex-specific effects on contextual and cued fear conditioning in adulthood. Physiol Behav 2021; 231:113314. [PMID: 33417904 DOI: 10.1016/j.physbeh.2021.113314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Stress-induced deviations in central nervous system development has long-term effects on adult mental health. Previous research in humans demonstrates that prenatal or adolescent stress increases the risk for psychiatric disorders. Animal models investigating the effects of stress during prenatal or adolescent development produces behavioral outcomes analogous to those observed in humans. However, whether adolescent stress exposure potentiates the effects of prenatal stress is currently unknown. Thus, the current study tested whether adolescent stress increases the impact of prenatal stress on contextual and cued fear memory in adulthood. Male and female Sprague Dawley rats were exposed to a chronic variable stress schedule during the last week of gestation, during adolescence, or during both developmental periods before undergoing fear conditioning training in adulthood. Our hypothesis predicted that the combined effects of prenatal and adolescent stress on contextual and cued fear memory would be greater than the effects of stress during either time period alone. In contrast to our hypothesis, however, we found independent effects of prenatal and adolescent stress on contextual and cued fear memory in both sexes, with no additional combined impact of stress exposure during both developmental phases. In males, developmental stress increased freezing behavior during contextual and cued testing regardless of whether stress exposure was prenatal, adolescent, or combined prenatal and adolescent stress exposure. In contrast, the effects of developmental stress in females were both test- and ovarian hormone status-dependent. During cued testing, nonstressed female freezing behavior depended on estrous cycle phase, whereas freezing behavior in stressed females did not, suggesting that developmental stress interferes with hormone-dependent cued fear memory. No effects of developmental stress or estrous cycle phase were observed for contextual fear memory in females. The results of the current study suggest that the effects of prenatal and adolescent stress on contextual and cued fear memory are not cumulative, but the effects of developmental stress on associative memory differ between males and females.
Collapse
|
33
|
Lisi G, Ribolsi M, Siracusano A, Niolu C. Maternal Vitamin D and its Role in Determining Fetal Origins of Mental Health. Curr Pharm Des 2020; 26:2497-2509. [PMID: 32370709 DOI: 10.2174/1381612826666200506093858] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/26/2020] [Indexed: 12/21/2022]
Abstract
There is evidence that mental health disorders may have roots in fetal life and are associated with deficiencies in various micronutrients, including vitamin D. During pregnancy, vitamin D balance is influenced by an increase in maternal calcitriol and a substantial increase in maternal Vitamin D Binding Protein concentrations. In the early stages of life, vitamin D is necessary to mediate numerous brain processes such as proliferation, apoptosis, and neurotransmission. Furthermore, Vitamin D has a recognized anti-inflammatory activity that normally suppresses inflammation. Increased activation of hypothalamo-pituitary-adrenal axis (HPA) and inflammation during gestation may influence maternal health and fetal neurodevelopment during and beyond pregnancy. A deficit of Vitamin D and maternal stressful events during gestation, such as perinatal depression, could influence the efficacy of the immune system altering its activity. Vitamin D deficiency during gestation associated with a reduction in fetal brain development has been widely described and correlated with alteration in the production of the brain-derived neurotrophic factor. To this regard, many studies highlights that low maternal vitamin D dosage during gestation has been related to a significantly greater risk to develop schizophrenia and other severe mental illnesses in later life. The objective of this paper is a comprehensive overview of maternal vitamin D balance in determining the fetal origins of mental health with some references to the link between vitamin D levels, inflammatory responses to stress and mental disorders in adult life.
Collapse
Affiliation(s)
- Giulia Lisi
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Mental Health, ASL ROMA 1, Rome, Italy
| | - Michele Ribolsi
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Policlinico Tor Vergata Foundation, Rome, Italy
| | - Alberto Siracusano
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Policlinico Tor Vergata Foundation, Rome, Italy
| | - Cinzia Niolu
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Policlinico Tor Vergata Foundation, Rome, Italy
| |
Collapse
|
34
|
Gyllenberg D, McKeague IW, Sourander A, Brown AS. Robust data-driven identification of risk factors and their interactions: A simulation and a study of parental and demographic risk factors for schizophrenia. Int J Methods Psychiatr Res 2020; 29:1-11. [PMID: 32520440 PMCID: PMC7723216 DOI: 10.1002/mpr.1834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/12/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Few interactions between risk factors for schizophrenia have been replicated, but fitting all such interactions is difficult due to high-dimensionality. Our aims are to examine significant main and interaction effects for schizophrenia and the performance of our approach using simulated data. METHODS We apply the machine learning technique elastic net to a high-dimensional logistic regression model to produce a sparse set of predictors, and then assess the significance of odds ratios (OR) with Bonferroni-corrected p-values and confidence intervals (CI). We introduce a simulation model that resembles a Finnish nested case-control study of schizophrenia which uses national registers to identify cases (n = 1,468) and controls (n = 2,975). The predictors include nine sociodemographic factors and all interactions (31 predictors). RESULTS In the simulation, interactions with OR = 3 and prevalence = 4% were identified with <5% false positive rate and ≥80% power. None of the studied interactions were significantly associated with schizophrenia, but main effects of parental psychosis (OR = 5.2, CI 2.9-9.7; p < .001), urbanicity (1.3, 1.1-1.7; p = .001), and paternal age ≥35 (1.3, 1.004-1.6; p = .04) were significant. CONCLUSIONS We have provided an analytic pipeline for data-driven identification of main and interaction effects in case-control data. We identified highly replicated main effects for schizophrenia, but no interactions.
Collapse
Affiliation(s)
- David Gyllenberg
- Department of Child Psychiatry, University of Turku, Turku, Finland.,Department of Adolescent Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.,Welfare Department, National Institute for Health and Welfare, Helsinki, Finland
| | - Ian W McKeague
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Andre Sourander
- Department of Child Psychiatry, University of Turku, Turku, Finland.,Department of Child Psychiatry, Turku University Central Hospital, Turku, Finland.,Department of Psychiatry, College of Physicians and Surgeons of Columbia University and New York State Psychiatric Institute, New York, New York, USA
| | - Alan S Brown
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University and New York State Psychiatric Institute, New York, New York, USA.,Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| |
Collapse
|
35
|
Fitzgerald E, Hor K, Drake AJ. Maternal influences on fetal brain development: The role of nutrition, infection and stress, and the potential for intergenerational consequences. Early Hum Dev 2020; 150:105190. [PMID: 32948364 PMCID: PMC7481314 DOI: 10.1016/j.earlhumdev.2020.105190] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An optimal early life environment is crucial for ensuring ideal neurodevelopmental outcomes. Brain development consists of a finely tuned series of spatially and temporally constrained events, which may be affected by exposure to a sub-optimal intra-uterine environment. Evidence suggests brain development may be particularly vulnerable to factors such as maternal nutrition, infection and stress during pregnancy. In this review, we discuss how maternal factors such as these can affect brain development and outcome in offspring, and we also identify evidence which suggests that the outcome can, in many cases, be stratified by socio-economic status (SES), with individuals in lower brackets typically having a worse outcome. We consider the relevant epidemiological evidence and draw parallels to mechanisms suggested by preclinical work where appropriate. We also discuss possible transgenerational effects of these maternal factors and the potential mechanisms involved. We conclude that modifiable factors such as maternal nutrition, infection and stress are important contributors to atypical brain development and that SES also likely has a key role.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kahyee Hor
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Amanda J Drake
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
36
|
Prenatal developmental origins of behavior and mental health: The influence of maternal stress in pregnancy. Neurosci Biobehav Rev 2020; 117:26-64. [DOI: 10.1016/j.neubiorev.2017.07.003] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 04/09/2017] [Accepted: 07/11/2017] [Indexed: 01/17/2023]
|
37
|
Dong E, Pandey SC. Prenatal stress induced chromatin remodeling and risk of psychopathology in adulthood. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 156:185-215. [PMID: 33461663 PMCID: PMC7864549 DOI: 10.1016/bs.irn.2020.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
New insights into the pathophysiology of psychiatric disorders suggest the existence of a complex interplay between genetics and environment. This notion is supported by evidence suggesting that exposure to stress during pregnancy exerts profound effects on the neurodevelopment and behavior of the offspring and predisposes them to psychiatric disorders later in life. Accumulated evidence suggests that vulnerability to psychiatric disorders may result from permanent negative effects of long-term changes in synaptic plasticity due to altered epigenetic mechanisms (histone modifications and DNA methylation) that lead to condensed chromatin architecture, thereby decreasing the expression of candidate genes during early brain development. In this chapter, we have summarized the literature of clinical studies on psychiatric disorders induced by maternal stress during pregnancy. We also discussed the epigenetic alterations of gene regulations induced by prenatal stress. Because the clinical manifestations of psychiatric disorders are complex, it is obvious that the biological progression of these diseases cannot be studied only in postmortem brains of patients and the use of animal models is required. Therefore, in this chapter, we have introduced a well-established mouse model of prenatal stress (PRS) generated in restrained pregnant dams. The behavioral phenotypes of the offspring (PRS mice) born to the stressed dam and underlying epigenetic changes in key molecules related to synaptic activity were described and highlighted. PRS mice may serve as a useful model for investigating the pathogenesis of psychiatric disorders and may be a useful tool for screening for the potential compounds that may normalize aberrant epigenetic mechanisms induced by prenatal stress.
Collapse
Affiliation(s)
- Erbo Dong
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
38
|
Kwiatkowski MA, Cope ZA, Lavadia ML, van de Cappelle CJA, Dulcis D, Young JW. Short-active photoperiod gestation induces psychiatry-relevant behavior in healthy mice but a resiliency to such effects are seen in mice with reduced dopamine transporter expression. Sci Rep 2020; 10:10217. [PMID: 32576854 PMCID: PMC7311429 DOI: 10.1038/s41598-020-66873-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/13/2020] [Indexed: 01/02/2023] Open
Abstract
A higher incidence of multiple psychiatric disorders occurs in people born in late winter/early spring. Reduced light exposure/activity level impacts adult rodent behavior and neural mechanisms, yet few studies have investigated such light exposure on gestating fetuses. A dysfunctional dopamine system is implicated in most psychiatric disorders, and genetic polymorphisms reducing expression of the dopamine transporter (DAT) are associated with some conditions. Furthermore, adult mice with reduced DAT expression (DAT-HT) were hypersensitive to short active (SA; 19:5 L:D) photoperiod exposure versus their wildtype (WT) littermates. Effects of SA photoperiod exposure during gestation in these mice have not been examined. We confirmed adult females exhibit a heightened corticosterone response when in SA photoperiod. We then tested DAT-HT mice and WT littermates in psychiatry-relevant behavioral tests after SA or normal active (NA; 12:12 L:D) photoperiod exposure during gestation and early life. SA-born WT mice exhibited sensorimotor gating deficits (males), increased reward preference, less immobility, open arm avoidance (females), less motivation to obtain a reward, and reversal learning deficits, vs. NA-born WT mice. DAT-HT mice were largely resilient to these effects, however. Future studies will determine the mechanism(s) by which SA photoperiod exposure influences brain development to predispose toward emergence of psychiatry-relevant behaviors.
Collapse
Affiliation(s)
- Molly A Kwiatkowski
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Zackary A Cope
- Department of Medicine, Aging Institute, University of Pittsburgh, Pittsburgh, USA
| | - Maria L Lavadia
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Chuck J A van de Cappelle
- Department of Psychiatry, University of California, San Diego, San Diego, USA.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Davide Dulcis
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Jared W Young
- Department of Psychiatry, University of California, San Diego, San Diego, USA. .,Research Service, VA San Diego Healthcare System, San Diego, USA.
| |
Collapse
|
39
|
Sex differences in the interactive effects of early life stress and the endocannabinoid system. Neurotoxicol Teratol 2020; 80:106893. [PMID: 32437941 DOI: 10.1016/j.ntt.2020.106893] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022]
Abstract
Sex differences in both the endocannabinoid system and stress responses have been established for decades. While there is ample evidence that the sexes respond differently to stress and that the endocannabinoid system is involved in this response, what is less clear is whether the endocannabinoid system mediates this response to stress differently in both sexes. Also, do the sexes respond similarly to exogenous cannabinoids (CBs) following stress? Can the administration of exogenous CBs normalize the effects of stress and if so, does this happen similarly in male and female subjects? This review will attempt to delineate the stress induced neurochemical alterations in the endocannabinoid system and the resulting behavioral changes across periods of development: prenatal, early neonatal or adolescent in males and females. Within this frame work, we will then examine the neurochemical and behavioral effects of exogenous CBs and illustrate that the response to CBs is determined by the stress history of the animal. The theoretical framework for this endeavor relates to the established effects of adverse childhood experiences (ACE) in increasing substance abuse, depression and anxiety and the possibility that individuals with high ACE scores may consume cannabinoids to "self-medicate". Overall, we see that while there are instances where exogenous cannabinoids "normalize" the adverse effects produced by early stress, this normalization does not occur in all animal models with any sort of consistency. The most compelling report where CB administration appears to normalize behaviors altered by early stress, shows minimal differences between the sexes (Alteba et al., 2016). This is in stark contrast to the majority of studies on early stress and the endocannabinoid system where both sexes are included and show quite divergent, in fact opposite, effects in males and females. Frequently there is a disconnect between neurochemical changes and behavioral changes and often, exogenous CBs have greater effects in stressed animals compared to non-stressed controls. This report as well as others reviewed here do support the concept that the effects of exogenous CBs are different in individuals experiencing early stress and that these differences are not equal in males and females. However, due to the wide variety of stressors used and the range of ages when the stress is applied, additional careful studies are warranted to fully understand the interactive effects of stress and the endocannabinoid system in males and females. In general, the findings do not support the statement that CB self-administration is an effective treatment for the adverse behavioral effects of early maltreatment in either males or females. Certainly this review should draw the attention of clinicians working with children, adolescents and adults exposed to early trauma and provide some perspective on the dysregulation of the endocannabinoid system in the response to trauma, the complex actions of exogenous CBs based on stress history and the unique effects of these factors in men and women.
Collapse
|
40
|
Lu Y, Jiang J, Si J, Wu Q, Tian F, Jiao K, Mu Y, Dong P, Zhu Z. PDLIM5 improves depression-like behavior of prenatal stress offspring rats via methylation in male, but not female. Psychoneuroendocrinology 2020; 115:104629. [PMID: 32171900 DOI: 10.1016/j.psyneuen.2020.104629] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Prenatal stress (PS) contributes to depression-like behavior in the offspring. PDLIM5 is involved in the onset of mental disorders. This study is to investigate the role and mechanism of PDLIM5 in depression-like behavior of PS offspring rats. METHODS PS model was used to analyze the effects of different treatments to PS offspring rats with different sex, including PDLIM5, PDLIM5 shRNA and 5-aza-2' -deoxycytidine (5-azaD). The depression-like behavior was assessed by the sucrose preference test (SPT) and forced swimming test (FST). The mRNA and protein expression levels of PDLIM5 in the hippocampus of PS offspring rats were detected by qRT-PCR and western blot, respectively. The methylation of PDLIM5 promoter were analyzed by bisulfite sequencing. RESULTS Our data revealed that PS offspring rats showed a significant decrease in sucrose preference and a prolonged immobility time. Injection of PDLIM5 significantly improved the depression-like behavior in PS offspring rats, whereas administration of PDLIM5 shRNA aggravated it. In addition, PDLIM5 expression was decreased at the mRNA and protein levels, and the methylation level of PDLIM5 promoter was increased in hippocampus of PS male but not female offspring rats. Furthermore, microinjection of 5-azaD improved the PS induced depression-like behavior in offspring rats. Moreover, in male PS offspring rats, microinjection of 5-azaD reversed the effect of PS on PDLIM5 expression and promoter methylation. CONCLUSION PDLIM5 can significantly improve the depression-like behavior of both male and female PS offspring rats, while the PDLIM5 promoter methylation is only observed in male PS offspring rats. Our study may provide new mechanism for the pathogenesis of depression and experimental evidence for sex-based precise treatment.
Collapse
Affiliation(s)
- Yong Lu
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Jiguo Jiang
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Jingfang Si
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Qi Wu
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Fengjuan Tian
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Keling Jiao
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Yingjun Mu
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Peng Dong
- Central Laboratory, Heze Medical College, Heze, 274000, China
| | - Zhongliang Zhu
- Maternal and Infant Health Research Institute and Medical College, Northwestern University, Xi'an, 710069, China.
| |
Collapse
|
41
|
De Matteis T, D'Andrea G, Lal J, Berardi D, Tarricone I. The impact of peri-natal stress on psychosis risk: results from the Bo-FEP incidence study. BMC Res Notes 2020; 13:153. [PMID: 32178713 PMCID: PMC7074980 DOI: 10.1186/s13104-020-04992-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/03/2020] [Indexed: 11/12/2022] Open
Abstract
Objective According to the gene-environment interaction model the pathogenesis of psychosis relies on an adverse neuro-socio-developmental pathway. Perinatal stress represents an important risk factor for the development of psychosis because of the increasingly evident interference with socio-neuro-development in the earlier phases of life. We aim to investigate the correlation of perinatal risk factors with the onset of psychosis with a case–control–incidence study. Results Patients (and their mothers) were eligible if they presented with first-episode psychosis at the Bologna West Community Mental Health Centre (Bo-West CMHC) between 2002 and 2012. The Bo-West CMHC serves a catchment area of about 200,000 people. The controls were recruited in the same catchment area and study period. 42 patients, 26 controls and their mothers were included. We collected the history of peri-natal stress and calculated crude and adjusted Odds Ratios for onset of first-episode psychosis. Adjusted logistic regression showed that psychosis onset was significantly associated with stressful situations during pregnancy, lower level of maternal physical health before or during pregnancy, use of anti-inflammatory drugs during pregnancy, and low level of maternal education. The results of our study suggest that stress during perinatal period increases the risk of developing psychosis.
Collapse
Affiliation(s)
- Tiziano De Matteis
- Bologna Transcultural Psychosomatic Team (BoTPT), Department of Medical and Surgical Sciences, Alma Mater Studiorum Bologna University, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Bologna University, Via Giovanni Masserenti, 9 - Pavillon 11, 40138, Bologna, Italy
| | - Giuseppe D'Andrea
- Bologna Transcultural Psychosomatic Team (BoTPT), Department of Medical and Surgical Sciences, Alma Mater Studiorum Bologna University, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Bologna University, Via Giovanni Masserenti, 9 - Pavillon 11, 40138, Bologna, Italy
| | - Jatin Lal
- Bologna Transcultural Psychosomatic Team (BoTPT), Department of Medical and Surgical Sciences, Alma Mater Studiorum Bologna University, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Bologna University, Via Giovanni Masserenti, 9 - Pavillon 11, 40138, Bologna, Italy
| | - Domenico Berardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Bologna University, Via Giovanni Masserenti, 9 - Pavillon 11, 40138, Bologna, Italy
| | - Ilaria Tarricone
- Bologna Transcultural Psychosomatic Team (BoTPT), Department of Medical and Surgical Sciences, Alma Mater Studiorum Bologna University, Bologna, Italy. .,Department of Medical and Surgical Sciences (DIMEC), University of Bologna; Clinica Medica, Policlinico Sant'Orsola-Malpighi, Via Masserenti 9, 40138, Bologna, Italy.
| |
Collapse
|
42
|
Ferreira F, Castro D, Araújo AS, Fonseca AR, Ferreira TB. Exposure to Traumatic Events and Development of Psychotic Symptoms in a Prison Population: A Network Analysis Approach. Psychiatry Res 2020; 286:112894. [PMID: 32151849 DOI: 10.1016/j.psychres.2020.112894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 01/03/2023]
Abstract
Previous studies consistently observed an association between exposure to traumatic events and psychotic symptoms. However, little is known about the differential impact of distinct traumatic events and the role of general symptoms in mediating this relationship. Thus, our study aimed to explore the differential association of several traumatic events to the psychotic symptoms in a sample of prisoners and whether this association is mediated by general symptoms. The total sample from the Survey of Psychiatric Morbidity Among Prisoners in England and Wales (N = 3039; 75.4% male) was used. Participants completed a list of traumatic events experienced before reclusion, the Psychosis Screening Questionnaire, Clinical Review Schedule-Revised. Network analysis was used to estimate the network of interactions between traumatic events and general and psychotic symptoms. Shortest paths analysis was performed to identify the different development trajectories. Results suggested that memory problems, compulsions, and irritability might be key mediating symptoms for most traumatic events. However, sexual abuse showed alternative mediators that might be specific of this traumatic event. Finally, the traumatic events, suffered from violence at work, separation/divorce and been homeless showed direct associations with specific psychotic symptoms.
Collapse
Affiliation(s)
- Filipa Ferreira
- University Institute of Maia, Avenida Carlos Oliveira Campos Castêlo da Maia, 4475-690, Maia, Portugal; Center for Psychology at University of Porto.
| | - Daniel Castro
- University Institute of Maia, Avenida Carlos Oliveira Campos Castêlo da Maia, 4475-690, Maia, Portugal; Center for Psychology at University of Porto
| | - Ana Sofia Araújo
- University Institute of Maia, Avenida Carlos Oliveira Campos Castêlo da Maia, 4475-690, Maia, Portugal
| | - Ana Rita Fonseca
- University Institute of Maia, Avenida Carlos Oliveira Campos Castêlo da Maia, 4475-690, Maia, Portugal
| | - Tiago Bento Ferreira
- University Institute of Maia, Avenida Carlos Oliveira Campos Castêlo da Maia, 4475-690, Maia, Portugal; Center for Psychology at University of Porto
| |
Collapse
|
43
|
Campbell RK, Tamayo-Ortiz M, Cantoral A, Schnaas L, Osorio-Valencia E, Wright RJ, Téllez-Rojo MM, Wright RO. Maternal Prenatal Psychosocial Stress and Prepregnancy BMI Associations with Fetal Iron Status. Curr Dev Nutr 2020; 4:nzaa018. [PMID: 32099952 PMCID: PMC7026381 DOI: 10.1093/cdn/nzaa018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Iron accrued in utero is critical for fetal and infant neurocognitive development. Psychosocial stress and obesity can each suppress fetal iron accrual. Their combined effects and differences by fetal sex are not known. In an observational pregnancy cohort study in Mexico City, we investigated associations of maternal prenatal life stressors, psychological dysfunction, and prepregnancy BMI with fetal iron status at delivery. OBJECTIVES We hypothesized that greater maternal prenatal psychosocial stress and prepregnancy overweight and obesity are associated with lower cord blood ferritin and hemoglobin (Hb), with stronger associations in boys than girls. METHODS Psychosocial stress in multiple domains of life stress (negative life events, perceived stress, exposure to violence) and psychological dysfunction symptoms (depression, generalized anxiety, and pregnancy-specific anxiety) were assessed with validated questionnaires during pregnancy. Prepregnancy BMI was predicted with a validated equation and categorized as normal/overweight/obese. Cord blood ferritin and Hb associations with prenatal psychosocial stress and BMI were modeled in multivariable linear regressions adjusted for maternal age, socioeconomic status, child sex, and prenatal iron supplementation. Interactions with child sex and 3-way stress-overweight/obesity-sex interactions were tested with product terms and likelihood ratio tests. RESULTS In 493 dyads, median (IQR) cord blood ferritin and Hb concentrations were 185 µg/L (126-263 g/dL) and 16 g/dL (14.7-17.1 g/dL), respectively. Ferritin was lower in infants of mothers with higher prenatal perceived stress (-23%; 95% CI: -35%, -9%), violence exposure (-28%; 95% CI: -42%, -12%), anxiety symptoms (-16%; 95% CI: -27%, -4%), and obesity (-17%; 95% CI: -31%, 0.2%). Interaction models suggested sex differences and synergism between maternal stress and overweight/obesity. No associations were observed between stress or BMI and Hb. CONCLUSIONS Multiple prenatal psychosocial stressors and excess prepregnancy BMI were each inversely associated with fetal iron status at birth. Pregnancies and infants at elevated risk of impaired fetal iron accrual may be identifiable according to observed synergism between maternal stress and obesity and differential associations with fetal iron status by infant sex.
Collapse
Affiliation(s)
- Rebecca K Campbell
- Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcela Tamayo-Ortiz
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
- National Council for Science and Technology, Mexico City, Mexico
| | - Alejandra Cantoral
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
- National Council for Science and Technology, Mexico City, Mexico
| | - Lourdes Schnaas
- Division of Research in Community Interventions, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - Erika Osorio-Valencia
- Division of Research in Community Interventions, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - Rosalind J Wright
- Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martha M Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
44
|
Shaw JC, Crombie GK, Zakar T, Palliser HK, Hirst JJ. Perinatal compromise contributes to programming of GABAergic and glutamatergic systems leading to long-term effects on offspring behaviour. J Neuroendocrinol 2020; 32:e12814. [PMID: 31758712 DOI: 10.1111/jne.12814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/30/2019] [Accepted: 11/20/2019] [Indexed: 01/01/2023]
Abstract
Extensive evidence now shows that adversity during the perinatal period is a significant risk factor for the development of neurodevelopmental disorders long after the causative event. Despite stemming from a variety of causes, perinatal compromise appears to have similar effects on the developing brain, thereby resulting in behavioural disorders of a similar nature. These behavioural disorders occur in a sex-dependent manner, with males affected more by externalising behaviours such as attention deficit hyperactivity disorder (ADHD) and females by internalising behaviours such as anxiety. Regardless of the causative event or the sex of the offspring, these disorders may begin in childhood or adolescence but extend into adulthood. A mechanism by which adverse events in the perinatal period impact later in life behaviour has been shown to be the changing epigenetic landscape. Methylation of the GAD1/GAD67 gene, which encodes the key glutamate-to-GABA-synthesising enzyme glutamate decarboxylase 1, resulting in increased levels of glutamate, is one epigenetic mechanism that may account for a tendency towards excitation in disorders such as ADHD. Exposure of the fetus or the neonate to high levels of cortisol may be the mediator between perinatal compromise and poor behavioural outcomes because evidence suggests that increased glucocorticoid exposure triggers widespread changes in the epigenetic landscape. This review summarises the current evidence and recent literature about the impact of various perinatal insults on the epigenome and the common mechanisms that may explain the similarity of behavioural outcomes occurring following diverse perinatal compromise.
Collapse
Affiliation(s)
- Julia C Shaw
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Gabrielle K Crombie
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tamas Zakar
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
45
|
Biskup E, Martinkova J, Ferretti MT. Gender medicine: Towards a gender-specific treatment of neuropsychiatric disorders. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:437-448. [PMID: 33008542 DOI: 10.1016/b978-0-444-64123-6.00029-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sex and gender are increasingly recognized as major influencing factors in disorders across all medical specialties. Even though there is ample evidence of sex and gender differences in neuropsychiatric disorders, a sex and gender-differentiated approach has not yet been sufficiently applied to diagnostics and management. Therefore, there is an urgent need to establish general recommendations and guidelines toward precision and sex/gender medicine, with regard to dosage, tolerability, interactions and side effects, sensitivity of diagnostic tests, and distinct treatment strategies. This chapter illustrates the current knowledge about sex and gender aspects in neuropsychiatric disorders, providing a base not only to assist the clinician in the handling of specific pathologic entities, but also to sensitize medical practitioners to consider sex and gender in clinical decision-making. As such, the chapter is a call to action to physicians and researchers to produce more sex- and gender-stratified evidence, leading to an acceleration of guideline development. Such novel guidelines will provide a base for medical education, of both medical students and specialists, as well as a reference point for practitioners, toward precision medicine.
Collapse
Affiliation(s)
- Ewelina Biskup
- Women's Brain Project, Guntershausen (TG), Switzerland; Shanghai University of Medicine and Health Sciences, College of Clinical Medicine, Shanghai, China.
| | - Julie Martinkova
- Women's Brain Project, Guntershausen (TG), Switzerland; Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | | |
Collapse
|
46
|
McQuaid GA, Darcey VL, Avalos MF, Fishbein DH, VanMeter JW. Altered cortical structure and psychiatric symptom risk in adolescents exposed to maternal stress in utero: A retrospective investigation. Behav Brain Res 2019; 375:112145. [PMID: 31400378 PMCID: PMC10561894 DOI: 10.1016/j.bbr.2019.112145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Maternal exposure to stress during pregnancy is associated with increased risk for cognitive and behavioral sequelae in offspring. Animal research demonstrates exposure to stress during gestation has effects on brain structure. In humans, however, little is known about the enduring effects of in utero exposure to maternal stress on brain morphology. We examine whether maternal report of stressful events during pregnancy is associated with brain structure and behavior in adolescents. We compare gray matter morphometry of typically-developing early adolescents (11-14 years of age, mean 12.7) at a single timepoint, based on presence/absence of retrospectively-assessed maternal report of negative major life event stress (MLES) during pregnancy: prenatal stress (PS; n = 28), comparison group (CG; n = 55). The Drug Use Screening Inventory Revised (DUSI-R) assessed adolescent risk for problematic behaviors. Exclusionary criteria included pre-term birth, low birth weight, and maternal substance use during pregnancy. Groups were equivalent for demographic (age, sex, IQ, SES, race/ethnicity), and birth measures (weight, length). Compared to CG peers, adolescents in the PS group exhibited increased gray matter density in bilateral posterior parietal cortex (PPC): bilateral intraparietal sulcus, left superior parietal lobule and inferior parietal lobule. Additionally, the PS group displayed greater risk for psychiatric symptoms and family system dysfunction, as assessed via DUSI-R subscales. These preliminary findings suggest that prenatal exposure to maternal MLES may exact enduring associations on offspring brain morphology and psychiatric risk, highlighting the importance of capturing these data in prospective longitudinal research studies (beginning at birth) to elucidate these associations.
Collapse
Affiliation(s)
- Goldie A McQuaid
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA.
| | - Valerie L Darcey
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA; Interdisciplinary Program in Neuroscience, Georgetown University, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| | - Melissa F Avalos
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| | - Diana H Fishbein
- Department of Human Development and Family Studies, Pennsylvania State University, 218 HHD Building, University Park, PA, 16802, USA
| | - John W VanMeter
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| |
Collapse
|
47
|
Ellman LM, Murphy SK, Maxwell SD, Calvo EM, Cooper T, Schaefer CA, Bresnahan MA, Susser ES, Brown AS. Maternal cortisol during pregnancy and offspring schizophrenia: Influence of fetal sex and timing of exposure. Schizophr Res 2019; 213:15-22. [PMID: 31345704 PMCID: PMC7074891 DOI: 10.1016/j.schres.2019.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Maternal stress during pregnancy has been repeatedly linked to increased risk for schizophrenia; however, no study has examined maternal cortisol during pregnancy and risk for the disorder. Study aims were to determine whether prenatal cortisol was associated with risk for schizophrenia and risk for an intermediate phenotype-decreased fetal growth-previously linked to prenatal cortisol and schizophrenia. Timing of exposure and fetal sex also were examined given previous findings. METHODS Participants were 64 cases diagnosed with schizophrenia spectrum disorders (SSD) and 117 controls from a prospective birth cohort study. Maternal cortisol was determined from stored sera from each trimester and psychiatric diagnoses were assessed from offspring using semi-structured interviews and medical records review. RESULTS Maternal cortisol during pregnancy was not associated with risk for offspring schizophrenia. There was a significant interaction between 3rd trimester cortisol and case status on fetal growth. Specifically, cases exposed to higher 3rd trimester maternal cortisol had significantly decreased fetal growth compared to controls. In addition, these findings were restricted to male offspring. CONCLUSIONS Our results indicate that higher prenatal cortisol is associated with an intermediate phenotype linked to schizophrenia, fetal growth, but only among male offspring who developed schizophrenia. Findings were consistent with evidence that schizophrenia genes may disrupt placental functioning specifically for male fetuses, as well as findings that males are more vulnerable to maternal cortisol during pregnancy. Finally, results suggest that examining fetal sex and intermediate phenotypes may be important in understanding the mechanisms involved in prenatal contributors to schizophrenia.
Collapse
Affiliation(s)
- Lauren M Ellman
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Shannon K Murphy
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Seth D Maxwell
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Evan M Calvo
- Department of Psychology, Temple University, Weiss Hall, 1701 N. 13(th) Street, Philadelphia, PA 19106, United States of America.
| | - Thomas Cooper
- Analytic Psychopharmacology, Nathan S. Kline Institute, 140 Old Orangeburg Road Orangeburg, NY 10962, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America; Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Catherine A Schaefer
- Division of Research, Kaiser Permanente, 2000 Broadway, Oakland, CA 94612, United States of America.
| | - Michaeline A Bresnahan
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Ezra S Susser
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America.
| | - Alan S Brown
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168(th) Street, New York, NY 10032, United States of America; New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States of America; Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States of America.
| |
Collapse
|
48
|
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: 17.0] [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]
|
49
|
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: 42] [Impact Index Per Article: 7.0] [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
|
50
|
Verstraeten BSE, McCreary JK, Falkenberg EA, Fang X, Weyers S, Metz GAS, Olson DM. Multiple prenatal stresses increase sexual dimorphism in adult offspring behavior. Psychoneuroendocrinology 2019; 107:251-260. [PMID: 31174163 DOI: 10.1016/j.psyneuen.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Maternal gestational stress and immune activation have independently been associated with affective and neurodevelopmental disorders across the lifespan. We investigated whether rats exposed to prenatal maternal stressors (PNMS) consisting of psychological stress, interleukin (IL)-1β or both (two-hit stress) during critical developmental windows displayed a behavioral phenotype representative of these conditions. METHODS Long-Evans dams were exposed to psychological stressors consisting of restraint stress and forced swimming from gestational day (GD)12 to 18 or to no stress (controls). From GD17 until day of delivery, these same animals were injected with saline or IL-1β as a second hit and immune stressor (5 μg/day, intraperitoneally). The behavior of F1 offspring adults was tested on the open field test, elevated plus maze and affective exploration task on postnatal days (P)90, 100 and 110 respectively. RESULTS The effects of PNMS differed depending on the specific testing environment and potentially the age at assessment, especially in female offspring. Both locomotion and anxiety-like behavioral measures were susceptible to PNMS effects. In females, psychological stress increased anxiety-like behavior, whereas IL-1β had an opposite effect, inducing exploration and risk-taking behavior on the open field test and the elevated plus maze. When present, interactions between both stressors limited the anxiogenic effect of psychological stress on its own. In contrast, prenatal psychological stress increased anxiety-like behavior in adult males overall. A similar anxiogenic effect of IL-1β was only found on the open field test while the Stress*IL-1β interaction appeared to limit the effect of either alone. Contrarily, the PNMS effects on anxiety-like behavior on the affective exploration task were highly similar between both sexes. Analysis of males and females together revealed an additive effect of Stress and IL-1β on the number of exits from the refuge, a measure of risk assessment and thus correlated with anxiety. CONCLUSION PNMS affected offspring adult behavior in a sex-dependent manner. Effects on females were more variable, whereas psychological stress mostly induced anxiety-like behavior in males. These data highlight the sexual dimorphism in vulnerability to prenatal stressors. Maternal or stress-induced programming of the stress response and neuroinflammation may play an important role in mediating stress effects on offspring adult behavior.
Collapse
Affiliation(s)
- Barbara S E Verstraeten
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada; Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - J Keiko McCreary
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Erin A Falkenberg
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Xin Fang
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada
| | - Steven Weyers
- Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada.
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|