151
|
Abstract
Stress is associated with the onset of several stress-related mental disorders that occur more frequently in women than in men, such as major depression or posttraumatic stress disorder (PTSD). The hypothalamic-pituitary-adrenal (HPA) axis is the major component of the neuroendocrine network responding to internal and external challenges. The proper functioning of the HPA axis is critical for the maintenance of mental and physical health, as dysregulations of the HPA axis have been linked to several mental and physical disorders. Numerous studies have observed distinct sex differences in the regulation of the HPA axis in response to stress, and it is supposed that these differences may partially explain the female predominance in stress-related mental disorders. Preclinical models have clearly shown that the HPA axis in females is activated more rapidly and produces a larger output of stress hormones than in males. However, studies with humans often produced inconsistent findings, which might be traced back to the variation of investigated stressors, the use of contraceptives in some of the studies, and different menstrual cycle stages of the female subjects. This article discusses rodent and human literature of sex differences in the function of the HPA axis.
Collapse
Affiliation(s)
- Carolin Leistner
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany; Comprehensive Heart Failure Center, University Hospital of Wuerzburg, Wuerzburg, Germany; Department of Psychosomatic Medicine and Psychotherapy, Medical Park Chiemseeblick, Bernau-Felden, Germany.
| |
Collapse
|
152
|
Jiménez Vásquez FDJ, Guerrero DM, Osornio MR, Rubio Osornio MDC, Suárez SO, Retana-Márquez S. Decreased serotonin content and release in the ventral hippocampus of prenatally stressed male rats in response to forced swim test. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
153
|
|
154
|
Chen MX, Liu Q, Cheng S, Lei L, Lin AJ, Wei R, K Hui TC, Li Q, Ao LJ, Sham PC. Interleukin-18 levels in the hippocampus and behavior of adult rat offspring exposed to prenatal restraint stress during early and late pregnancy. Neural Regen Res 2020; 15:1748-1756. [PMID: 32209782 PMCID: PMC7437598 DOI: 10.4103/1673-5374.276358] [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] [Indexed: 12/17/2022] Open
Abstract
Exposure to maternal stress during prenatal life is associated with an increased risk of neuropsychiatric disorders, such as depression and anxiety, in offspring. It has also been increasingly observed that prenatal stress alters the phenotype of offspring via immunological mechanisms and that immunological dysfunction, such as elevated interleukin-18 levels, has been reported in cultures of microglia. Prenatal restraint stress (PRS) in rats permits direct experimental investigation of the link between prenatal stress and adverse outcomes. However, the majority of studies have focused on the consequences of PRS delivered in the second half of pregnancy, while the effects of early prenatal stress have rarely been examined. Therefore, pregnant rats were subjected to PRS during early/middle and late gestation (days 8-14 and 15-21, respectively). PRS comprised restraint in a round plastic transparent cylinder under bright light (6500 lx) three times per day for 45 minutes. Differences in interleukin-18 expression in the hippocampus and in behavior were compared between offspring rats and control rats on postnatal day 75. We found that adult male offspring exposed to PRS during their late prenatal periods had higher levels of anxiety-related behavior and depression than control rats, and both male and female offspring exhibited higher levels of depression-related behavior, impaired recognition memory and diminished exploration of novel objects. Moreover, an elevated level of interleukin-18 was observed in the dorsal and ventral hippocampus of male and female early- and late-PRS offspring rats. The results indicate that PRS can cause anxiety and depression-related behaviors in adult offspring and affect the expression of interleukin-18 in the hippocampus. Thus, behavior and the molecular biology of the brain are affected by the timing of PRS exposure and the sex of the offspring. All experiments were approved by the Animal Experimentation Ethics Committee at Kunming Medical University, China (approval No. KMMU2019074) in January 2019.
Collapse
Affiliation(s)
- Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shu Cheng
- Department of Rehabilitation, China Resources & WISCO General Hospital, Wuhan, Hubei Province, China
| | - Lei Lei
- Department of Rehabilitation Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ai-Jin Lin
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Ran Wei
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Tomy C K Hui
- Department of Psychiatry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qi Li
- Department of Psychiatry; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Pak C Sham
- Department of Psychiatry; State Key Laboratory of Brain and Cognitive Sciences; Centre for Genomic Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
155
|
Widen EM, Nichols AR, Kahn LG, Factor-Litvak P, Insel BJ, Hoepner L, Dube SM, Rauh V, Perera F, Rundle A. Prepregnancy obesity is associated with cognitive outcomes in boys in a low-income, multiethnic birth cohort. BMC Pediatr 2019; 19:507. [PMID: 31862007 PMCID: PMC6924019 DOI: 10.1186/s12887-019-1853-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/22/2019] [Indexed: 01/13/2023] Open
Abstract
Background Maternal obesity and high gestational weight gain (GWG) disproportionally affect low-income populations and may be associated with child neurodevelopment in a sex-specific manner. We examined sex-specific associations between prepregnancy BMI, GWG, and child neurodevelopment at age 7. Methods Data are from a prospective low-income cohort of African American and Dominican women (n = 368; 44.8% male offspring) enrolled during the second half of pregnancy from 1998 to 2006. Neurodevelopment was measured using the Wechsler Intelligence Scale for Children (WISC-IV) at approximately child age 7. Linear regression estimated associations between prepregnancy BMI, GWG, and child outcomes, adjusting for race/ethnicity, marital status, gestational age at delivery, maternal education, maternal IQ and child age. Results Overweight affected 23.9% of mothers and obesity affected 22.6%. At age 7, full-scale IQ was higher among girls (99.7 ± 11.6) compared to boys (96.9 ± 13.3). Among boys, but not girls, prepregnancy overweight and obesity were associated with lower full-scale IQ scores [overweight β: − 7.1, 95% CI: (− 12.1, − 2.0); obesity β: − 5.7, 95% CI: (− 10.7, − 0.7)]. GWG was not associated with full-scale IQ in either sex. Conclusions Prepregnancy overweight and obesity were associated with lower IQ among boys, but not girls, at 7 years. These findings are important considering overweight and obesity prevalence and the long-term implications of early cognitive development.
Collapse
Affiliation(s)
- Elizabeth M Widen
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA. .,Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.
| | - Amy R Nichols
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA.,Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA
| | - Linda G Kahn
- Department of Pediatrics, New York University School of Medicine, 403 East 34th St, New York, NY, 10016, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| | - Beverly J Insel
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Lori Hoepner
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA.,Department of Environmental and Occupational Health Sciences, SUNY Downstate Medical Center, School of Public Health, 450 Clarkson Avenue, MSC 43, Brooklyn, NY, 11203, USA
| | - Sara M Dube
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA.,Department of Nutritional Sciences, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Virginia Rauh
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, 60 Haven Avenue, B-2, Room 213, New York, NY, 10032, USA
| | - Frederica Perera
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| | - Andrew Rundle
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| |
Collapse
|
156
|
Siller SJ, Rubenstein DR. A Tissue Comparison of DNA Methylation of the Glucocorticoid Receptor Gene (Nr3c1) in European Starlings. Integr Comp Biol 2019; 59:264-272. [PMID: 31076777 DOI: 10.1093/icb/icz034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Negative feedback of the vertebrate stress response via the hypothalamic-pituitary-adrenal (HPA) axis is regulated by glucocorticoid receptors in the brain. Epigenetic modification of the glucocorticoid receptor gene (Nr3c1), including DNA methylation of the promoter region, can influence expression of these receptors, impacting behavior, physiology, and fitness. However, we still know little about the long-term effects of these modifications on fitness. To better understand these fitness effects, we must first develop a non-lethal method to assess DNA methylation in the brain that allows for multiple measurements throughout an organism's lifetime. In this study, we aimed to determine if blood is a viable biomarker for Nr3c1 DNA methylation in two brain regions (hippocampus and hypothalamus) in adult European starlings (Sturnus vulgaris). We found that DNA methylation of CpG sites in the complete Nr3c1 putative promoter varied among tissue types and was lowest in blood. Although we identified a similar cluster of correlated Nr3c1 putative promoter CpG sites within each tissue, this cluster did not show any correlation in DNA methylation among tissues. Additional studies should consider the role of the developmental environment in producing epigenetic modifications in different tissues.
Collapse
Affiliation(s)
- Stefanie J Siller
- Department of Ecology, Evolution and Environmental Biology, Columbia University, 10th Floor Schermerhorn Extension, 1200 Amsterdam Avenue, New York, NY, USA
| | - Dustin R Rubenstein
- Department of Ecology, Evolution and Environmental Biology, Columbia University, 10th Floor Schermerhorn Extension, 1200 Amsterdam Avenue, New York, NY, USA
| |
Collapse
|
157
|
Sze Y, Brunton PJ. Sex, stress and steroids. Eur J Neurosci 2019; 52:2487-2515. [DOI: 10.1111/ejn.14615] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
| | - Paula J. Brunton
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
- Zhejiang University‐University of Edinburgh Joint Institute Haining Zhejiang China
| |
Collapse
|
158
|
Tomasi J, Lisoway AJ, Zai CC, Harripaul R, Müller DJ, Zai GCM, McCabe RE, Richter MA, Kennedy JL, Tiwari AK. Towards precision medicine in generalized anxiety disorder: Review of genetics and pharmaco(epi)genetics. J Psychiatr Res 2019; 119:33-47. [PMID: 31563039 DOI: 10.1016/j.jpsychires.2019.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023]
Abstract
Generalized anxiety disorder (GAD) is a prevalent and chronic mental disorder that elicits widespread functional impairment. Given the high degree of non-response/partial response among patients with GAD to available pharmacological treatments, there is a strong need for novel approaches that can optimize outcomes, and lead to medications that are safer and more effective. Although investigations have identified interesting targets predicting treatment response through pharmacogenetics (PGx), pharmaco-epigenetics, and neuroimaging methods, these studies are often solitary, not replicated, and carry several limitations. This review provides an overview of the current status of GAD genetics and PGx and presents potential strategies to improve treatment response by combining better phenotyping with PGx and improved analytical methods. These strategies carry the dual benefit of delivering data on biomarkers of treatment response as well as pointing to disease mechanisms through the biology of the markers associated with response. Overall, these efforts can serve to identify clinical, genetic, and epigenetic factors that can be incorporated into a pharmaco(epi)genetic test that may ultimately improve treatment response and reduce the socioeconomic burden of GAD.
Collapse
Affiliation(s)
- Julia Tomasi
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Amanda J Lisoway
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Clement C Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ricardo Harripaul
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel J Müller
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gwyneth C M Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; General Adult Psychiatry and Health Systems Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Randi E McCabe
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Anxiety Treatment and Research Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| | - Margaret A Richter
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Frederick W. Thompson Anxiety Disorders Centre, Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - James L Kennedy
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Arun K Tiwari
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
159
|
Behura SK, Dhakal P, Kelleher AM, Balboula A, Patterson A, Spencer TE. The brain-placental axis: Therapeutic and pharmacological relevancy to pregnancy. Pharmacol Res 2019; 149:104468. [PMID: 31600597 PMCID: PMC6944055 DOI: 10.1016/j.phrs.2019.104468] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/23/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022]
Abstract
The placenta plays a critical role in mammalian reproduction. Although it is a transient organ, its function is indispensable to communication between the mother and fetus, and supply of nutrients and oxygen to the growing fetus. During pregnancy, the placenta is vulnerable to various intrinsic and extrinsic conditions which can result in increased risk of fetal neurodevelopmental disorders as well as fetal death. The placenta controls the neuroendocrine secretion in the brain as a means of adaptive processes to safeguard the fetus from adverse programs, to optimize fetal development and other physiological changes necessary for reproductive success. Although a wealth of information is available on neuroendocrine functions in pregnancy, they are largely limited to the regulation of hypothalamus-pituitary-adrenal/gonad (HPA/ HPG) axis, particularly the oxytocin and prolactin system. There is a major gap in knowledge on systems-level functional interaction between the brain and placenta. In this review, we aim to outline the current state of knowledge about the brain-placental axis with description of the functional interactions between the placenta and the maternal and fetal brain. While describing the brain-placental interactions, a special emphasis has been given on the therapeutics and pharmacology of the placental receptors to neuroligands expressed in the brain during gestation. As a key feature of this review, we outline the prospects of integrated pharmacogenomics, single-cell sequencing and organ-on-chip systems to foster priority areas in this field of research. Finally, we remark on the application of precision genomics approaches to study the brain-placental axis in order to accelerate personalized medicine and therapeutics to treat placental and fetal brain disorders.
Collapse
Affiliation(s)
- Susanta K Behura
- Division of Animal Sciences, University of Missouri, United States; Informatics Institute, University of Missouri, United States.
| | - Pramod Dhakal
- Division of Animal Sciences, University of Missouri, United States
| | | | - Ahmed Balboula
- Division of Animal Sciences, University of Missouri, United States
| | - Amanda Patterson
- Division of Animal Sciences, University of Missouri, United States; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, United States
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, United States; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, United States
| |
Collapse
|
160
|
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: 3.6] [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
|
161
|
Evidence of decreased gap junction coupling between astrocytes and oligodendrocytes in the anterior cingulate cortex of depressed suicides. Neuropsychopharmacology 2019; 44:2099-2111. [PMID: 31374562 PMCID: PMC6897926 DOI: 10.1038/s41386-019-0471-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Glial dysfunction is a major pathophysiological feature of mood disorders. While altered astrocyte (AS) and oligodendrocyte-lineage (OL) functions have been associated with depression, the crosstalk between these glial cell types has never been assessed in that context. AS are potent regulators of myelination, in part through gap junction (GJ) channels formed by the heterotypic coupling of AS-specific (Cx30 and Cx43) and OL-specific (Cx32 and Cx47) connexins. This study therefore aimed at addressing the integrity of AS/OL coupling in the anterior cingulate cortex (ACC) of depressed suicides. Using immunofluorescence and confocal imaging, we characterized the distribution of Cx30 and mapped its expression onto OL somas, myelinated axons, and brain vasculature in postmortem brain samples from depressed suicides (N = 48) and matched controls (N = 23). Differential gene expression of key components of the GJ nexus was also screened through RNA-sequencing previously generated by our group, and validated by quantitative real-time PCR. We show that Cx30 expression localized onto OL cells and myelinated fibers is decreased in deep cortical layers of the ACC in male-depressed suicides. This effect was associated with decreased expression of OL-specific connexins, as well as the downregulation of major connexin-interacting proteins essential for the scaffolding, trafficking, and function of GJs. These results provide a first evidence of impaired AS/OL GJ-mediated communication in the ACC of individuals with mood disorders. These changes in glial coupling are likely to have significant impact on brain function, and may contribute to the altered OL function previously reported in this brain region.
Collapse
|
162
|
Rosa MJ, Nentin F, Enlow MB, Hacker MR, Pollas N, Coull B, Wright RJ. Sex-specific associations between prenatal negative life events and birth outcomes. Stress 2019; 22:647-653. [PMID: 31057018 PMCID: PMC6776698 DOI: 10.1080/10253890.2019.1608944] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Maternal psychosocial stress can negatively impact gestational length and development of the fetus. These effects may be sex-specific but have not been extensively studied. The objective of this study was to examine the associations between prenatal maternal stress and birth outcomes and whether effects are modified by sex. Prenatal maternal stress was indexed by a maternal negative life events (NLEs) score ascertained in 527 urban mothers; a higher NLE score indicates greater stress. Birth outcomes included gestational age, preterm birth (PTB) (<37 weeks), and birthweight for gestational age z-scores. Modified Poisson regression and linear models were used to evaluate associations of prenatal NLE scores with birth outcomes. Sex differences were assessed by inclusion of an interaction term for sex by NLE score and in sex-stratified analyses. In analyses adjusted for maternal age, education, race/ethnicity, and pre-pregnancy body mass index (BMI), increasing prenatal stress was associated with shortened gestational age (days) (β = -0.63, [95% CI -1.20, -0.06]). This effect was sex specific, with increasing prenatal stress associated with shortened gestational age, as well as increased risk of PTB, in male infants (β = -1.35 [95% CI -2.17, -0.54] and RR = 1.18 [95% CI 0.99, 1.42], respectively) but not female infants (β = 0.15 [95%CI -0.63, 0.94] and RR = 0.85, [95%CI 0.65, 1.11], respectively). Prenatal stress was not associated with birthweight z-scores. Our results support the importance of psychosocial stress as a programming factor that may have sex-specific effects for adverse fetal outcomes. Understanding sex-specific effects of prenatal stress on birth outcomes may inform prevention strategies. LAY SUMMARY Higher stress experienced by mothers in pregnancy was associated with shorter length of pregnancy and the effect was stronger in male infants when compared to female infants.
Collapse
Affiliation(s)
- Maria José Rosa
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount, Sinai, New York, NY, USA
| | - Farida Nentin
- Department of Obstetrics, Gynecology and Reproductive Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Michele R. Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, KS3, Boston, MA 02215, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | | | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rosalind J. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount, Sinai, New York, NY, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
163
|
David I, Canario L, Combes S, Demars J. Intergenerational Transmission of Characters Through Genetics, Epigenetics, Microbiota, and Learning in Livestock. Front Genet 2019; 10:1058. [PMID: 31737041 PMCID: PMC6834772 DOI: 10.3389/fgene.2019.01058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Evolutionary biologists studying wild species have demonstrated that genetic and non-genetic sources of information are inherited across generations and are therefore responsible for phenotypic resemblance between relatives. Although it has been postulated that non-genetic sources of inheritance are important in natural selection, they are not taken into account for livestock selection that is based on genetic inheritance only. According to the natural selection theory, the contribution of non-genetic inheritance may be significant for the transmission of characters. If this theory is confirmed in livestock, not considering non-genetic means of transmission in selection schemes might prevent achieving maximum progress in the livestock populations being selected. The present discussion paper reviews the different mechanisms of genetic and non-genetic inheritance reported in the literature as occurring in livestock species. Non-genetic sources of inheritance comprise information transmitted via physical means, such as epigenetic and microbiota inheritance, and those transmitted via learning mechanisms: behavioral, cultural and ecological inheritance. In the first part of this paper we review the evidence that suggests that both genetic and non-genetic information contribute to inheritance in livestock (i.e. transmitted from one generation to the next and causing phenotypic differences between individuals) and discuss how the environment may influence non-genetic inherited factors. Then, in a second step, we consider methods for favoring the transmission of non-genetic inherited factors by estimating and selecting animals on their extended transmissible value and/or introducing favorable non-genetic factors via the animals’ environment.
Collapse
Affiliation(s)
- Ingrid David
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Laurianne Canario
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Julie Demars
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| |
Collapse
|
164
|
Polli FS, Kohlmeier KA. Prenatal Nicotine Exposure in Rodents: Why Are There So Many Variations in Behavioral Outcomes? Nicotine Tob Res 2019; 22:1694-1710. [DOI: 10.1093/ntr/ntz196] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/05/2019] [Indexed: 01/01/2023]
Abstract
Abstract
Introduction
The World Health Organization (WHO) reported that smoking cessation rates among women have stagnated in the past decade and estimates that hundreds of millions of women will be smokers in the next decade. Social, environmental, and biological conditions render women more susceptible to nicotine addiction, imposing additional challenges to quit smoking during gestation, which is likely why more than 8% of pregnancies in Europe are associated with smoking. In epidemiological investigations, individuals born from gestational exposure to smoking exhibit a higher risk of development of attention-deficit/hyperactive disorder (ADHD) and liability to drug dependence. Among other teratogenic compounds present in tobacco smoke, nicotine actions during neuronal development could contribute to the observed outcomes as nicotine misleads signaling among progenitor cells during brain development. Several experimental approaches have been developed to address the consequences of prenatal nicotine exposure (PNE) to the brain and behavior but, after four decades of studies, inconsistent data have been reported and the lack of consensus in the field has compromised the hypothesis that gestational nicotine exposure participates in cognitive and emotional behavioral deficits.
Aims
In this review, we discuss the most commonly used PNE models with focus on their advantages and disadvantages, their relative validity, and how the different technical approaches could play a role in the disparate outcomes.
Results
We propose methodological considerations, which could improve the translational significance of the PNE models.
Conclusions
Such alterations might be helpful in reconciling experimental findings, as well as leading to development of treatment targets for maladaptive behaviors in those prenatally exposed.
Implications
In this article, we have reviewed the advantages and disadvantages of different variables of the commonly used experimental models of PNE. We discuss how variations in the nicotine administration methods, the timing of nicotine exposure, nicotine doses, and species employed could contribute to the disparate findings in outcomes for PNE offspring, both in behavior and neuronal changes. In addition, recent findings suggest consideration of epigenetic effects extending across generations. Finally, we have suggested improvements in the available PNE models that could contribute to the enhancement of their validity, which could assist in the reconciliation of experimental findings.
Collapse
Affiliation(s)
- Filip Souza Polli
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristi Anne Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
165
|
Nolvi S, Bridgett DJ, Korja R, Kataja EL, Junttila N, Karlsson H, Karlsson L. Trajectories of maternal pre- and postnatal anxiety and depressive symptoms and infant fear: Moderation by infant sex. J Affect Disord 2019; 257:589-597. [PMID: 31330484 DOI: 10.1016/j.jad.2019.07.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/05/2019] [Accepted: 07/04/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Prior work has examined the links between pre- and postnatal maternal distress and infant negative affectivity; however, there is little understanding about how the continuity of infant exposure to pre- and postnatal maternal distress relates to infant development. This study investigated the continuity of maternal pre- and postnatal depressive and anxiety symptoms and their relations with infant fear among 391 mother-infant dyads. An additional aim was to consider infant sex as a moderating factor. METHODS Maternal anxiety and depressive symptoms were measured during gestational weeks 14, 24 and 34 and 3 and 6 months postpartum. Subsequently, infant fear was measured using mother reports (IBQ-R) at 6 months and in a laboratory setting (Lab-TAB Masks episode) at 8 months. Using growth mixture modeling, a three-class model describing the course of maternal symptoms across pregnancy and the early postnatal period was identified, consisting of mothers with "Consistently Low Distress", "Prenatal-Only Distress", and "Consistently High Distress". RESULTS Infant girls exposed to prenatal-only maternal distress were higher in observed fear than infant boys exposed to prenatal-only distress. Infant girls exposed to consistently high distress also showed lower observed fear than their counterparts exposed to prenatal-only maternal distress. LIMITATIONS The main limitation of the study is the relatively small group size within the Consistently High subgroup. CONCLUSIONS The findings suggest that girls might be particularly sensitive to maternal distress, and that prenatal-only and continuous distress exposure are differentially related to female infant fear.
Collapse
Affiliation(s)
- Saara Nolvi
- FinnBrain Birth Cohort Study, Department of Clinical Medicine, Turku Brain and Mind Center, University of Turku, Turku, Finland; Department of Medical Psychology, Charité - Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany..
| | - David J Bridgett
- Department of Psychology, Northern Illinois University, DeKalb, IL, United States
| | - Riikka Korja
- FinnBrain Birth Cohort Study, Department of Clinical Medicine, Turku Brain and Mind Center, University of Turku, Turku, Finland; Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Eeva-Leena Kataja
- FinnBrain Birth Cohort Study, Department of Clinical Medicine, Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Niina Junttila
- Department of Teacher Education, University of Turku, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Department of Clinical Medicine, Turku Brain and Mind Center, University of Turku, Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Department of Clinical Medicine, Turku Brain and Mind Center, University of Turku, Turku, Finland; Department of Child Psychiatry, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
166
|
Williams S, Ghosh C. Neurovascular glucocorticoid receptors and glucocorticoids: implications in health, neurological disorders and drug therapy. Drug Discov Today 2019; 25:89-106. [PMID: 31541713 DOI: 10.1016/j.drudis.2019.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/12/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
Glucocorticoid receptors (GRs) are ubiquitous transcription factors widely studied for their role in controlling events related to inflammation, stress and homeostasis. Recently, GRs have reemerged as crucial targets of investigation in neurological disorders, with a focus on pharmacological strategies to direct complex mechanistic GR regulation and improve therapy. In the brain, GRs control functions necessary for neurovascular integrity, including responses to stress, neurological changes mediated by the hypothalamic-pituitary-adrenal axis and brain-specific responses to corticosteroids. Therefore, this review will examine GR regulation at the neurovascular interface in normal and pathological conditions, pharmacological GR modulation and glucocorticoid insensitivity in neurological disorders.
Collapse
Affiliation(s)
- Sherice Williams
- Brain Physiology Laboratory/Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chaitali Ghosh
- Brain Physiology Laboratory/Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine and Biomedical Engineering at Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
167
|
Hodes GE, Epperson CN. Sex Differences in Vulnerability and Resilience to Stress Across the Life Span. Biol Psychiatry 2019; 86:421-432. [PMID: 31221426 PMCID: PMC8630768 DOI: 10.1016/j.biopsych.2019.04.028] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/08/2019] [Accepted: 04/25/2019] [Indexed: 12/12/2022]
Abstract
Susceptibility and resilience to stress depend on 1) the timing of the exposure with respect to development, 2) the time across the life span at which effects are measured, and 3) the behavioral or biological phenotype under consideration. This translational review examines preclinical stress models that provide clues to causal mechanisms and their relationship to the more complex phenomenon of stress-related psychiatric and cognitive disorders in humans. We examine how genetic sex and epigenetic regulation of hormones contribute to the proximal and distal effects of stress at different epochs of life. Stress during the prenatal period and early postnatal life puts male offspring at risk of developing diseases involving socialization, such as autism spectrum disorder, and attention and cognition, such as attention-deficit/hyperactivity disorder. While female offspring show resilience to some of the proximal effects of prenatal and early postnatal stress, there is evidence that risk associated with developmental insults is unmasked in female offspring following periods of hormonal activation and flux, including puberty, pregnancy, and perimenopause. Likewise, stress exposures during puberty have stronger proximal effects on girls, including an increased risk of developing mood-related and stress-related illnesses, such as depression, anxiety, and posttraumatic stress disorder. Hormonal changes during menopause and andropause impact the processes of memory and emotion in women and men, though women are preferentially at risk for dementia, and childhood adversity further impacts estradiol effects on neural function. We propose that studies to determine mechanisms for stress risk and resilience across the life span must consider the nature and timing of stress exposures as well as the sex of the organism under investigation.
Collapse
Affiliation(s)
- Georgia E. Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - C. Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
168
|
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: 1.6] [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
|
169
|
Tahira AC, Barbosa AR, Feltrin AS, Gastaldi VD, de Toledo VHC, de Carvalho Pereira JG, Lisboa BCG, de Souza Reis VN, dos Santos ACF, Maschietto M, Brentani H. Putative contributions of the sex chromosome proteins SOX3 and SRY to neurodevelopmental disorders. Am J Med Genet B Neuropsychiatr Genet 2019; 180:390-414. [PMID: 30537354 PMCID: PMC6767407 DOI: 10.1002/ajmg.b.32704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022]
Abstract
The male-biased prevalence of certain neurodevelopmental disorders and the sex-biased outcomes associated with stress exposure during gestation have been previously described. Here, we hypothesized that genes distinctively targeted by only one or both homologous proteins highly conserved across therian mammals, SOX3 and SRY, could induce sexual adaptive changes that result in a differential risk for neurodevelopmental disorders. ChIP-seq/chip data showed that SOX3/SRY gene targets were expressed in different brain cell types in mice. We used orthologous human genes in rodent genomes to extend the number of SOX3/SRY set (1,721). These genes were later found to be enriched in five modules of coexpressed genes during the early and mid-gestation periods (FDR < 0.05), independent of sexual hormones. Genes with differential expression (24, p < 0.0001) and methylation (40, p < 0.047) between sexes were overrepresented in this set. Exclusive SOX3 or SRY target genes were more associated with the late gestational and postnatal periods. Using autism as a model sex-biased disorder, the SOX3/SRY set was enriched in autism gene databases (FDR ≤ 0.05), and there were more de novo variations from the male autism spectrum disorder (ASD) samples under the SRY peaks compared to the random peaks (p < 0.024). The comparison of coexpressed networks of SOX3/SRY target genes between male autism and control samples revealed low preservation in gene modules related to stress response (99 genes) and neurogenesis (78 genes). This study provides evidence that while SOX3 is a regulatory mechanism for both sexes, the male-exclusive SRY also plays a role in gene regulation, suggesting a potential mechanism for sex bias in ASD.
Collapse
Affiliation(s)
- Ana Carolina Tahira
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - André Rocha Barbosa
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
- Inter‐institutional Grad Program on BioinformaticsUniversity of São PauloSão PauloSPBrazil
| | | | - Vinicius Daguano Gastaldi
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Victor Hugo Calegari de Toledo
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | | | - Bianca Cristina Garcia Lisboa
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Viviane Neri de Souza Reis
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Ana Cecília Feio dos Santos
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
- Laboratório de Pesquisas Básicas em Malária – EntomologiaSeção de Parasitologia – Instituto Evandro Chagas/SVS/MSAnanindeuaPABrazil
| | - Mariana Maschietto
- Brazilian Biosciences National Laboratory (LNBio)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSPBrazil
| | - Helena Brentani
- LIM23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
- Inter‐institutional Grad Program on BioinformaticsUniversity of São PauloSão PauloSPBrazil
- Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSPBrazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD)Sao PauloSPBrazil
- Faculdade de Medicina FMUSPUniversidade de Sao PauloSao PauloSPBrazil
| |
Collapse
|
170
|
Keller SM, Nowak A, Roth TL. Female pups receive more maltreatment from stressed dams. Dev Psychobiol 2019; 61:824-831. [PMID: 30810229 PMCID: PMC6711830 DOI: 10.1002/dev.21834] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 01/10/2023]
Abstract
The effects of exposure to developmental stress often diverge for males and females. Using the scarcity-adversity model of low nesting resources outside the home cage, our lab has discovered sex differences in both behavioral and epigenetic consequences of repeated exposure to caregiver maltreatment. For the measures we have performed to date, we have found more consequences for females. The reasons underlying this sex disparity are unknown. In the current experiment, we aimed to discern the quality of maternal care received by male and female pups in our model. As we have previously found more behavioral and epigenetic consequences in females, we hypothesized that females receive more adverse care compared to their male littermates. Our hypothesis was supported; in our maltreatment condition, we found that female pups received more adverse care than males. This sex difference in adverse care was not present in our two control conditions (cross-foster and normal maternal care). These data lend support to the notion that one reason females in our model incur more behavioral and epigenetic consequences is a result of greater mistreatment by the dam.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Anna Nowak
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
171
|
Molecular programs underlying differences in the expression of mood disorders in males and females. Brain Res 2019; 1719:89-103. [DOI: 10.1016/j.brainres.2019.05.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/20/2019] [Accepted: 05/13/2019] [Indexed: 01/13/2023]
|
172
|
Tuscher JJ, Day JJ. Multigenerational epigenetic inheritance: One step forward, two generations back. Neurobiol Dis 2019; 132:104591. [PMID: 31470104 DOI: 10.1016/j.nbd.2019.104591] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/22/2019] [Accepted: 08/26/2019] [Indexed: 02/08/2023] Open
Abstract
Modifications to DNA and histone proteins serve a critical regulatory role in the developing and adult brain, and over a decade of research has established the importance of these "epigenetic" modifications in a wide variety of brain functions across the lifespan. Epigenetic patterns orchestrate gene expression programs that establish the phenotypic diversity of various cellular classes in the central nervous system, play a key role in experience-dependent gene regulation in the adult brain, and are commonly implicated in neurodevelopmental, psychiatric, and neurodegenerative disease states. In addition to these established roles, emerging evidence indicates that epigenetic information can potentially be transmitted to offspring, giving rise to inter- and trans-generational epigenetic inheritance phenotypes. However, our understanding of the cellular events that participate in this information transfer is incomplete, and the ability of this transfer to overcome complete epigenetic reprogramming during embryonic development is highly controversial. This review explores the existing literature on multigenerational epigenetic mechanisms in the central nervous system. First, we focus on the cellular mechanisms that may perpetuate or counteract this type of information transfer, and consider how epigenetic modification in germline and somatic cells regulate important aspects of cellular and organismal development. Next, we review the potential phenotypes resulting from ancestral experiences that impact gene regulatory modifications, including how these changes may give rise to unique metabolic phenotypes. Finally, we discuss several caveats and technical limitations that influence multigenerational epigenetic effects. We argue that studies reporting multigenerational epigenetic changes impacting the central nervous system must be interpreted with caution, and provide suggestions for how epigenetic information transfer can be mechanistically disentangled from genetic and environmental influences on brain function.
Collapse
Affiliation(s)
- Jennifer J Tuscher
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Jeremy J Day
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
173
|
Placental programming of neuropsychiatric disease. Pediatr Res 2019; 86:157-164. [PMID: 31003234 DOI: 10.1038/s41390-019-0405-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022]
Abstract
The placenta is vital for fetal growth, and compromised function is associated with abnormal development, especially of the brain. Linking placental function to brain development is a new field we have dubbed neuroplacentology. Approximately 380,000 infants in the United States each year abruptly lose placental support upon premature birth, and more than 10% of pregnancies are affected by more insidious placental dysfunction such as preeclampsia or infection. Abnormal fetal brain development or injury can lead to life-long neurological impairments, including psychiatric disorders. The majority of research connecting placental compromise to fetal brain injury has focused on gas exchange or nutritional programming, neglecting the placenta's essential neuroendocrine role. We will review the current evidence that placental dysfunction, particularly endocrine dysfunction, secretion of pro-inflammatory cytokines, or barrier breakdown may place many thousands of fetuses at risk for life-long neurodevelopmental impairments each year. Understanding how specific placental factors shape brain development and increase the risk for later psychiatric disorders, including autism, attention deficit disorder, and schizophrenia, paves the way for novel treatment strategies to maintain the normal developmental milieu and protect from further injury.
Collapse
|
174
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
175
|
Velasquez JC, Zhao Q, Chan Y, Galindo LC, Simasotchi C, Wu D, Hou Z, Herod SM, Oberlander TF, Gil S, Fournier T, Burd I, Andrews AM, Bonnin A. In Utero Exposure to Citalopram Mitigates Maternal Stress Effects on Fetal Brain Development. ACS Chem Neurosci 2019; 10:3307-3317. [PMID: 31184110 DOI: 10.1021/acschemneuro.9b00180] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Human epidemiological and animal-model studies suggest that separate exposure to stress or serotonin-selective reuptake inhibitor (SSRI) antidepressants during pregnancy increases risks for neurodevelopmental disorders in offspring. Yet, little is known about the combined effects of maternal stress and SSRIs with regard to brain development in utero. We found that the placenta is highly permeable to the commonly prescribed SSRI (±)-citalopram (CIT) in humans and mice, allowing rapid exposure of the fetal brain to this drug. We investigated the effects of maternal chronic unpredictable stress in mice with or without maternal oral administration of CIT from embryonic day (E)8 to E17. We assessed fetal brain development using magnetic resonance imaging and quantified changes in serotonergic, thalamocortical, and cortical development. In utero exposure to maternal stress did not affect overall fetal brain growth. However, serotonin tissue content in the fetal forebrain was increased in association with maternal stress; this increase was reversed by maternal CIT. In utero exposure to stress increased the numbers of deep-layer neurons in specific cortical regions, whereas CIT increased overall cell numbers without changing the proportions of layer-specific neurons to offset the effects of stress on deep-layer cortical development. These findings suggest that stress and SSRI exposure in utero differentially impact serotonin-dependent fetal neurodevelopment such that CIT reverses key effects of maternal gestational stress on offspring brain development.
Collapse
Affiliation(s)
- Juan C. Velasquez
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Qiuying Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Yen Chan
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Ligia C.M. Galindo
- Department of Anatomy, Federal University of Pernambuco, Recife 50670, Brazil
| | | | - Dan Wu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Zhipeng Hou
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Skyla M. Herod
- Department of Biology and Chemistry, Azusa Pacific University, Azusa, California 91702, United States
| | - Tim F. Oberlander
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3 V4, Canada
| | - Sophie Gil
- UMR-S 1139 INSERM/University of Paris Descartes, 75006 Paris, France
- PremUp Foundation, 75014 Paris, France
| | - Thierry Fournier
- UMR-S 1139 INSERM/University of Paris Descartes, 75006 Paris, France
- PremUp Foundation, 75014 Paris, France
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Anne M. Andrews
- Terry and Jane Semel Institute for Neuroscience & Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, and Departments of Psychiatry and Biobehavioral Sciences and Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Alexandre Bonnin
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
176
|
Zhu Y, Chen H, Zhao X, Li B, He H, Cheng H, Wang H, Ao Y. Decreased H3K9ac level of KLF4 mediates podocyte developmental toxicity induced by prenatal caffeine exposure in male offspring rats. Toxicol Lett 2019; 314:63-74. [PMID: 31306741 DOI: 10.1016/j.toxlet.2019.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/30/2019] [Accepted: 07/04/2019] [Indexed: 01/03/2023]
Abstract
This study aimed to verify the toxic effects of prenatal caffeine exposure (PCE) on the podocyte development in male offspring, and to explore the underlying intrauterine programming mechanisms. The pregnant rats were administered with caffeine (30 to 120 mg/kg⋅d) during gestational day (GD) 9 to 20. The male fetus on GD20 and the offspring at postnatal week (PW) 6 and PW28 were sacrificed. The results indicated that PCE caused ultrastructural abnormalities on podocyte, and inhibited the expression of podocyte marker genes such as Nephrin, Wilms tumor 1 (WT1), the histone 3 lysine 9 acetylation (H3K9ac) level in the Kruppel-like factor 4 (KLF4) promoter and its expression in the male offspring from GD20 to PW28. Meanwhile, the expression of glucocorticoid receptor (GR) and histone deacetylase 7 (HDAC7) in the fetus were increased by PCE. In vitro, corticosterone increased GR and HDAC7 whereas reduced the H3K9ac level of KLF4 and KLF4/Nephrin expression. KLF4 over-expression reversed the reduction of Nephrin expression, knockdown of HDAC7 and GR antagonist RU486 partially reversed the inhibitory effects of corticosterone on H3K9ac level and KLF4 expression. In conclusion, PCE caused podocyte developmental toxicity in male offspring, which was associated with corticosterone-induced low-functional programming of KLF4 through GR/HDAC7/H3K9ac pathway.
Collapse
Affiliation(s)
- Yanan Zhu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Haiyun Chen
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Xiaoqi Zhao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Bin Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hangyuan He
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Cheng
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Ying Ao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China.
| |
Collapse
|
177
|
Dowell J, Elser BA, Schroeder RE, Stevens HE. Cellular stress mechanisms of prenatal maternal stress: Heat shock factors and oxidative stress. Neurosci Lett 2019; 709:134368. [PMID: 31299286 DOI: 10.1016/j.neulet.2019.134368] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/19/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022]
Abstract
Development of the brain prenatally is affected by maternal experience and exposure. Prenatal maternal psychological stress changes brain development and results in increased risk for neuropsychiatric disorders. In this review, multiple levels of prenatal stress mechanisms (offspring brain, placenta, and maternal physiology) are discussed and their intersection with cellular stress mechanisms explicated. Heat shock factors and oxidative stress are closely related to each other and converge with the inflammation, hormones, and cellular development that have been more deeply explored as the basis of prenatal stress risk. Increasing evidence implicates cellular stress mechanisms in neuropsychiatric disorders associated with prenatal stress including affective disorders, schizophrenia, and child-onset psychiatric disorders. Heat shock factors and oxidative stress also have links with the mechanisms involved in other kinds of prenatal stress including external exposures such as environmental toxicants and internal disruptions such as preeclampsia. Integrative understanding of developmental neurobiology with these cellular and physiological mechanisms is necessary to reduce risks and promote healthy brain development.
Collapse
Affiliation(s)
- Jonathan Dowell
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| | - Benjamin A Elser
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA.
| | - Rachel E Schroeder
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA.
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, Iowa City, IA, USA.
| |
Collapse
|
178
|
Joshi A, Page CE, Damante M, Dye CN, Haim A, Leuner B, Lenz KM. Sex differences in the effects of early life stress exposure on mast cells in the developing rat brain. Horm Behav 2019; 113:76-84. [PMID: 31054843 DOI: 10.1016/j.yhbeh.2019.04.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/12/2019] [Accepted: 04/26/2019] [Indexed: 12/16/2022]
Abstract
Early life stress leads to long lasting effects on behavior. Neuroimmune cells have been implicated as key mediators of experience-induced changes in brain and behavioral development, in that they are highly responsive to stress. Mast cells are one such type of neuroimmune cell, but little is known about their role in brain development or following early life stress. Here, we assessed the impact of three different early life stress exposure paradigms on mast cell dynamics in the developing brain of male and female rats, focusing on the hippocampus and hypothalamus, where most mast cells reside. We found that exposure to two weeks of chronic variable stress during gestation led to increased mast cell number and activation in the female offspring hypothalamus on the day of birth. Acute exposure to maternal separation stress on postnatal day (PN) 2 led to significant decreases in mast cells within the hypothalamus and hippocampus of females, but not males. In contrast, one week of exposure to brief daily maternal separation stress (e.g., handling), increased mast cell numbers in the female, but not male, hippocampus. We found significant sex differences in mast cell number and activation, including males having more mast cells than females in the hippocampus on the day of birth and males having significantly more degranulated mast cells on PN11. Thus, mast cells may be an unappreciated mediator of sex-specific brain development in response to early life perturbations.
Collapse
Affiliation(s)
- Aarohi Joshi
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Chloe E Page
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Mark Damante
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Courtney N Dye
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Achikam Haim
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
179
|
Matthews SG, McGowan PO. Developmental programming of the HPA axis and related behaviours: epigenetic mechanisms. J Endocrinol 2019; 242:T69-T79. [PMID: 30917340 DOI: 10.1530/joe-19-0057] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
It has been approximately 30 years since the seminal discoveries of David Barker and his colleagues, and research is beginning to unravel the mechanisms that underlie developmental programming. The early environment of the embryo, foetus and newborn have been clearly linked to altered hypothalamic-pituitary-adrenal (HPA) function and related behaviours through the juvenile period and into adulthood. A number of recent studies have shown that these effects can pass across multiple generations. The HPA axis is highly responsive to the environment, impacts both central and peripheral systems and is critical to health in a wide variety of contexts. Mechanistic studies in animals are linking early exposures to adversity with changes in gene regulatory mechanisms, including modifications of DNA methylation and altered levels of miRNA. Similar associations are emerging from recent human studies. These findings suggest that epigenetic mechanisms represent a fundamental link between adverse early environments and developmental programming of later disease. The underlying biological mechanisms that connect the perinatal environment with modified long-term health outcomes represent an intensive area of research. Indeed, opportunities for early interventions must identify the relevant environmental factors and their molecular targets. This new knowledge will likely assist in the identification of individuals who are at risk of developing poor outcomes and for whom early intervention is most effective.
Collapse
Affiliation(s)
- Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Departments of Obstetrics & Gynaecology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Patrick O McGowan
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, University of Toronto, Scarborough, Ontario, Canada
- Department of Cell and Systems Biology, Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
180
|
Saboory E, Mohammadi S, Dindarian S, Mohammadi H. Prenatal stress and elevated seizure susceptibility: Molecular inheritable changes. Epilepsy Behav 2019; 96:122-131. [PMID: 31132613 DOI: 10.1016/j.yebeh.2019.04.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/17/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023]
Abstract
Stressful episodes are common during early-life and may have a wide range of negative effects on both physical and mental status of the offspring. In addition to various neurobehavioral complications induced by prenatal stress (PS), seizure is a common complication with no fully explained cause. In this study, the association between PS and seizure susceptibility was reviewed focusing on sex differences and various underlying mechanisms. The role of drugs in the initiation of seizure and the effects of PS on the nervous system that prone the brain for seizure, especially the hypothalamic-pituitary-adrenal (HPA) axis, are also discussed in detail by reviewing the papers studying the effect of PS on glutamatergic, gamma-aminobutyric acid (GABA)ergic, and adrenergic systems in the context of seizure and epilepsy. Finally, epigenetic changes in epilepsy are described, and the underlying mechanisms of this change are expanded. As the effects of PS may be life-lasting, it is possible to prevent future psychiatric and behavioral disorders including epilepsy by preventing avoidable PS risk factors.
Collapse
Affiliation(s)
- Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Sedra Mohammadi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
| | - Sina Dindarian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hozan Mohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
181
|
Barrientos RM, Brunton PJ, Lenz KM, Pyter L, Spencer SJ. Neuroimmunology of the female brain across the lifespan: Plasticity to psychopathology. Brain Behav Immun 2019; 79:39-55. [PMID: 30872093 PMCID: PMC6591071 DOI: 10.1016/j.bbi.2019.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.
Collapse
Affiliation(s)
- R M Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH 43210, United States
| | - P J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK; Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang 314400, PR China
| | - K M Lenz
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychology, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, United States
| | - L Pyter
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic. 3083, Australia.
| |
Collapse
|
182
|
Mansell T, Ponsonby AL, Januar V, Novakovic B, Collier F, Burgner D, Vuillermin P, Ryan J, Saffery R. Early-life determinants of hypoxia-inducible factor 3A gene (HIF3A) methylation: a birth cohort study. Clin Epigenetics 2019; 11:96. [PMID: 31262346 PMCID: PMC6604333 DOI: 10.1186/s13148-019-0687-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/27/2019] [Indexed: 12/17/2022] Open
Abstract
Background Methylation of the hypoxia-inducible factor 3α gene (HIF3A) has been linked to pregnancy exposures, infant adiposity and later BMI. Genetic variation influences HIF3A methylation levels and may modify these relationships. However, data in very early life are limited, particularly in association with adverse pregnancy outcomes. We investigated the relationship between maternal and gestational factors, infant anthropometry, genetic variation and HIF3A DNA methylation in the Barwon Infant Study, a population-based birth cohort. Methylation of two previously studied regions of HIF3A were tested in the cord blood mononuclear cells of 938 infants. Results No compelling evidence was found of an association between birth weight, adiposity or maternal gestational diabetes with methylation at the most widely studied HIF3A region. Male sex (− 4.3%, p < 0.001) and pre-eclampsia (− 5.4%, p = 0.02) negatively associated with methylation at a second region of HIF3A; while positive associations were identified for gestational diabetes (4.8%, p = 0.01) and gestational age (1.2% increase per week, p < 0.001). HIF3A genetic variation also associated strongly with methylation at this region (p < 0.001). Conclusions Pre- and perinatal factors impact HIF3A methylation, including pre-eclampsia. This provides evidence that specific pregnancy complications, previously linked to adverse outcomes for both mother and child, impact the infant epigenome in a molecular pathway critical to several vascular and metabolic conditions. Further work is required to understand the mechanisms and clinical relevance, particularly the differing effects of in utero exposure to gestational diabetes or pre-eclampsia. Electronic supplementary material The online version of this article (10.1186/s13148-019-0687-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Toby Mansell
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Vania Januar
- Murdoch Children's Research Institute, Parkville, Australia
| | - Boris Novakovic
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Fiona Collier
- Murdoch Children's Research Institute, Parkville, Australia.,School of Medicine, Deakin University, Geelong, Australia.,Child Health Research Unit, Barwon Health, Geelong, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia.,Department of Paediatrics, Monash University, Clayton, Australia
| | - Peter Vuillermin
- Murdoch Children's Research Institute, Parkville, Australia.,School of Medicine, Deakin University, Geelong, Australia.,Child Health Research Unit, Barwon Health, Geelong, Australia
| | - Joanne Ryan
- Murdoch Children's Research Institute, Parkville, Australia.,School of Public Health & Preventive Medicine, Monash University, Melbourne, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Parkville, Australia. .,Department of Paediatrics, University of Melbourne, Parkville, Australia.
| | | |
Collapse
|
183
|
Early life stress and the propensity to develop addictive behaviors. Int J Dev Neurosci 2019; 78:156-169. [PMID: 31255718 DOI: 10.1016/j.ijdevneu.2019.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/03/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
There is a vast literature on effects of early life manipulations in rodents much of which is aimed at investigating the long-term consequences related to emotion and cognition in adulthood. Less is known about how these manipulations affect responses reflective of alcohol (AUD) and substance (SUD) use disorders. The purpose of this paper is to review the literature of studies that employed early life manipulations and assessed behavioral responses to psychoactive substances, specifically alcohol, opiates, and stimulants, in rodents. While the findings with alcohol are more limited and mixed, studies with opiates and stimulants show strong support for the ability of these manipulations to enhance behavioral responsivity to these substances in line with epidemiological data. Some outcomes show sex differences. The mechanisms that influence these enduring changes may reflect epigenetic alterations. Several studies support a role for altered DNA methylation (and other epigenetic mechanisms) as biological responses to early environmental insults. The chemical changes induced by DNA methylation affect transcriptional activity of DNA and thus can have a long-term impact on the individual's phenotype. Such effects are particularly robust when they occur during sensitive periods of brain development (e.g., first postnatal weeks in rodents). We review this emerging literature as it relates to the known neurobiology of AUDs and SUDs and suggest new avenues of research. Such findings will have implications for the treatment and prevention of AUDs and SUDs and could provide insight into factors that support resiliency.
Collapse
|
184
|
Raciti M, Salma J, Spulber S, Gaudenzi G, Khalajzeyqami Z, Conti M, Anderlid BM, Falk A, Hermanson O, Ceccatelli S. NRXN1 Deletion and Exposure to Methylmercury Increase Astrocyte Differentiation by Different Notch-Dependent Transcriptional Mechanisms. Front Genet 2019; 10:593. [PMID: 31316548 PMCID: PMC6610538 DOI: 10.3389/fgene.2019.00593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/05/2019] [Indexed: 01/11/2023] Open
Abstract
Controversial evidence points to a possible involvement of methylmercury (MeHg) in the etiopathogenesis of autism spectrum disorders (ASD). In the present study, we used human neuroepithelial stem cells from healthy donors and from an autistic patient bearing a bi-allelic deletion in the gene encoding for NRXN1 to evaluate whether MeHg would induce cellular changes comparable to those seen in cells derived from the ASD patient. In healthy cells, a subcytotoxic concentration of MeHg enhanced astroglial differentiation similarly to what observed in the diseased cells (N1), as shown by the number of GFAP positive cells and immunofluorescence signal intensity. In both healthy MeHg-treated and N1 untreated cells, aberrations in Notch pathway activity seemed to play a critical role in promoting the differentiation toward glia. Accordingly, treatment with the established Notch inhibitor DAPT reversed the altered differentiation. Although our data are not conclusive since only one of the genes involved in ASD is considered, the results provide novel evidence suggesting that developmental exposure to MeHg, even at subcytotoxic concentrations, induces alterations in astroglial differentiation similar to those observed in ASD.
Collapse
Affiliation(s)
- Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jahan Salma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Gaudenzi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Centre for Molecular Medicine, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
185
|
Buthmann J, Finik J, Ventura G, Zhang W, Shereen AD, Nomura Y. The children of Superstorm Sandy: Maternal prenatal depression blunts offspring electrodermal activity. Biol Psychol 2019; 146:107716. [PMID: 31176750 DOI: 10.1016/j.biopsycho.2019.107716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
We set out to examine the relations between prenatal exposure to the natural disaster Superstorm Sandy, maternal depression, and offspring electrodermal activity (EDA). EDA was measured via skin conductance response (SCR) magnitude in 198 children (M = 42.54 months, SD = 12.76) during a startle paradigm. In keeping with prior research, we expected prenatal depression to be associated with hyporeactive EDA and prenatal stress to be associated with hyperreactive EDA. SCR magnitude was lower in children prenatally exposed to depression alone, when compared to Superstorm Sandy, and controls. SCR magnitude of children prenatally exposed to both maternal depression and the storm was lower than that of all other groups. Our results emphasize the influence of maternal prenatal mental health, support targeted risk assessment for children who experienced an adverse prenatal environment, and highlight the need for a deeper understanding of the interactions between maternal mood and stress on the developing child.
Collapse
Affiliation(s)
- J Buthmann
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA; CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA.
| | - J Finik
- CUNY Graduate School of Public Health and Health Policy, Department of Epidemiology and Biostatistics, 55 W 125th St., New York, NY 10027, USA
| | - G Ventura
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - W Zhang
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA; New Jersey City University, Department of Psychology, 2039 John Fitzgerald Kennedy Blvd, Jersey City, NJ 07305, USA
| | - A D Shereen
- CUNY Advanced Science Research Center, Graduate Center, 85 St Nicholas Terrace, New York, NY 10031, USA; University of California, Department of Neurology, 680 California Ave, Irvine, CA 92697, USA
| | - Y Nomura
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA; CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA; CUNY Graduate School of Public Health and Health Policy, Department of Epidemiology and Biostatistics, 55 W 125th St., New York, NY 10027, USA; CUNY Advanced Science Research Center, Graduate Center, 85 St Nicholas Terrace, New York, NY 10031, USA; Mount Sinai School of Medicine, Department of Psychiatry, 1 Gustave Levy Pl, New York, NY 10029, USA
| |
Collapse
|
186
|
Polli A, Ickmans K, Godderis L, Nijs J. When Environment Meets Genetics: A Clinical Review of the Epigenetics of Pain, Psychological Factors, and Physical Activity. Arch Phys Med Rehabil 2019; 100:1153-1161. [DOI: 10.1016/j.apmr.2018.09.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023]
|
187
|
Conradt E, Ostlund B, Guerin D, Armstrong DA, Marsit CJ, Tronick E, LaGasse L, Lester BM. DNA methylation of NR3c1 in infancy: Associations between maternal caregiving and infant sex. Infant Ment Health J 2019; 40:513-522. [PMID: 31066465 DOI: 10.1002/imhj.21789] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Caregivers play a critical role in scaffolding infant stress reactivity and regulation, but the mechanisms by which this scaffolding occurs is unclear. Animal models strongly suggest that epigenetic processes, such as DNA methylation, are sensitive to caregiving behaviors and, in turn, offspring stress reactivity. We examined the direct effects of caregiving behaviors on DNA methylation in infants and infant stress reactivity. Infants and mothers (N = 128) were assessed during a free play when infants were 5 months old. Maternal responsiveness and appropriate touch were coded. and infant buccal epithelial cells were sampled to assess for DNA methylation of the glucocorticoid receptor gene, NR3c1 exon 1F. Infant cortisol reactivity was assessed in response to the still-face paradigm. Greater levels of maternal responsiveness and appropriate touch were related to less DNA methylation of specific regions in NR3c1 exon 1F, but only for females. There was no association with maternal responsiveness and appropriate touch or DNA methylation of NR3c1 exon 1F on prestress cortisol or cortisol reactivity. Our results are discussed in relation to programming models that implicate maternal care as an important factor in programing infant stress reactivity.
Collapse
Affiliation(s)
| | - Brendan Ostlund
- Department of Psychology, University of Utah, Salt Lake City, Utah
| | - Dylan Guerin
- Department of Pharmacology and Toxicology and of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David A Armstrong
- Department of Pharmacology and Toxicology and of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Carmen J Marsit
- Department of Pharmacology and Toxicology and of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Edward Tronick
- Department of Psychology, University of Massachusetts, Boston, Massachusetts
| | - Lyn LaGasse
- The Brown Center for the Study of Children at Risk, Providence, Rhode Island.,Department of Psychiatry, Warren Alpert Medical School of Brown University, Providence, Rhode Island.,Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Barry M Lester
- The Brown Center for the Study of Children at Risk, Providence, Rhode Island.,Department of Psychiatry, Warren Alpert Medical School of Brown University, Providence, Rhode Island.,Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
188
|
Monk C, Lugo-Candelas C, Trumpff C. Prenatal Developmental Origins of Future Psychopathology: Mechanisms and Pathways. Annu Rev Clin Psychol 2019; 15:317-344. [PMID: 30795695 PMCID: PMC7027196 DOI: 10.1146/annurev-clinpsy-050718-095539] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The developmental origins of health and disease hypothesis applied to neurodevelopmental outcomes asserts that the fetal origins of future development are relevant to mental health. There is a third pathway for the familial inheritance of risk for psychiatric illness beyond shared genes and the quality of parental care: the impact of pregnant women's distress-defined broadly to include perceived stress, life events, depression, and anxiety-on fetal and infant brain-behavior development. We discuss epidemiological and observational clinical data demonstrating that maternal distress is associated with children's increased risk for psychopathology: For example, high maternal anxiety is associated with a twofold increase in the risk of probable mental disorder in children. We review several biological systems hypothesized to be mechanisms by which maternal distress affects fetal and child brain and behavior development, as well as the clinical implications of studies of the developmental origins of health and disease that focus on maternal distress. Development and parenting begin before birth.
Collapse
Affiliation(s)
- Catherine Monk
- Department of Psychiatry, Columbia University, New York, NY 10032, USA;
- Department of Obstetrics and Gynecology, Columbia University, New York, NY 10032, USA
- New York State Psychiatric Institute, New York, NY 10032, USA; ,
| | - Claudia Lugo-Candelas
- Department of Psychiatry, Columbia University, New York, NY 10032, USA;
- New York State Psychiatric Institute, New York, NY 10032, USA; ,
| | - Caroline Trumpff
- Department of Psychiatry, Columbia University, New York, NY 10032, USA;
- New York State Psychiatric Institute, New York, NY 10032, USA; ,
| |
Collapse
|
189
|
Hamza M, Halayem S, Bourgou S, Daoud M, Charfi F, Belhadj A. Epigenetics and ADHD: Toward an Integrative Approach of the Disorder Pathogenesis. J Atten Disord 2019; 23:655-664. [PMID: 28665177 DOI: 10.1177/1087054717696769] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Epigenetic hypothesis is one of the research pathways used to explain the complex etiology of neurodevelopmental disorders. This review highlights the findings of recent studies in the field of epigenetics in ADHD. METHODS An electronic literature search using Medline. RESULTS In the Gene × Environment interaction model, several clinical, genetic and molecular arguments support the epigenetic hypothesis in ADHD etiology. Environmental ADHD risk factors including toxic, nutritional factors and stressful life events lead to changes in DNA methylation and in histone modification levels. One critical CpG site located in the promoter of the DRD4 gene exhibited a specific pattern in ADHD children. A methylome wide exploration of DNA showed decreased methylation in vasoactive intestinal peptide receptor 2 gene, which was not replicated by further research. CONCLUSION Current data require consolidation and could lead to the identification of biomarkers and the introduction of new modalities of treatment.
Collapse
Affiliation(s)
- Meriem Hamza
- 1 University of Tunis El Manar, Tunis, Tunisia.,2 Mongi Slim Hospital, Sidi Daoud, Tunisia
| | - Soumeyya Halayem
- 1 University of Tunis El Manar, Tunis, Tunisia.,3 Razi Hospital, Manouba, Tunisia
| | - Soumaya Bourgou
- 1 University of Tunis El Manar, Tunis, Tunisia.,2 Mongi Slim Hospital, Sidi Daoud, Tunisia
| | - Mona Daoud
- 1 University of Tunis El Manar, Tunis, Tunisia.,2 Mongi Slim Hospital, Sidi Daoud, Tunisia
| | - Fatma Charfi
- 1 University of Tunis El Manar, Tunis, Tunisia.,2 Mongi Slim Hospital, Sidi Daoud, Tunisia
| | - Ahlem Belhadj
- 1 University of Tunis El Manar, Tunis, Tunisia.,2 Mongi Slim Hospital, Sidi Daoud, Tunisia
| |
Collapse
|
190
|
Nelson LH, Saulsbery AI, Lenz KM. Small cells with big implications: Microglia and sex differences in brain development, plasticity and behavioral health. Prog Neurobiol 2019; 176:103-119. [PMID: 30193820 PMCID: PMC8008579 DOI: 10.1016/j.pneurobio.2018.09.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/17/2018] [Accepted: 09/01/2018] [Indexed: 12/20/2022]
Abstract
Brain sex differences are programmed largely by sex hormone secretions and direct sex chromosome effects in early life, and are subsequently modulated by early life experiences. The brain's resident immune cells, called microglia, actively contribute to brain development. Recent research has shown that microglia are sexually dimorphic, especially during early life, and may participate in sex-specific organization of the brain and behavior. Likewise, sex differences in immune cells and their signaling in the adult brain have been found, although in most cases their function remains unclear. Additionally, immune cells and their signaling have been implicated in many disorders in which brain development or plasticity is altered, including autism, schizophrenia, pain disorders, major depression, and postpartum depression. This review summarizes what is currently known about sex differences in neuroimmune function in development and during other major phases of brain plasticity, as well as the current state of knowledge regarding sex-specific neuroimmune function in psychiatric disorders.
Collapse
Affiliation(s)
- Lars H Nelson
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Angela I Saulsbery
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
191
|
Ancestral Stress Alters Lifetime Mental Health Trajectories and Cortical Neuromorphology via Epigenetic Regulation. Sci Rep 2019; 9:6389. [PMID: 31011159 PMCID: PMC6476877 DOI: 10.1038/s41598-019-42691-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/03/2019] [Indexed: 12/17/2022] Open
Abstract
Experiences during early development are powerful determinants of lifetime mental health. Here we investigated if ancestral stress regulates the brain’s epigenetic memory to alter neuromorphology and emotionality in the remote F4 progeny. Pregnant female rat dams of the parental F0 generation were exposed to stress on gestational days 12–18. To generate a transgenerational stress lineage, their pregnant daughters (F1), grand-daughters (F2) and great-grand-daughters (F3) remained undisturbed. To generate a multigenerational stress lineage, pregnant dams of each generation (F1–F3) were stressed. A lineage of non-stress controls (F0–F3) was also produced. Multigenerational stress exceeded the impact of transgenerational stress by increasing anxiety-like behaviours and stress response in young and middle-aged F4 males but not females. Functional changes were accompanied by reduced spine density in the male medial prefrontal cortex with opposite effects in the orbital frontal cortex. Ancestral stress regulated cortical miR-221 and miR-26 expression and their target genes, thus downregulating ntrk2 and map1a genes in males while downregulating crh and upregulating map1a genes in females. These miRNA-dependent pathways are candidates for developmental programming of lifetime mental health. Thus, multigenerational stress in particular determines sexually dimorphic predisposition to stress vulnerability and generates a phenotype resembling symptoms of post-traumatic stress disorder.
Collapse
|
192
|
Dantzer B, Dubuc C, Goncalves IB, Cram DL, Bennett NC, Ganswindt A, Heistermann M, Duncan C, Gaynor D, Clutton-Brock TH. The development of individual differences in cooperative behaviour: maternal glucocorticoid hormones alter helping behaviour of offspring in wild meerkats. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180117. [PMID: 30966876 PMCID: PMC6460081 DOI: 10.1098/rstb.2018.0117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2018] [Indexed: 01/04/2023] Open
Abstract
The phenotype of parents can have long-lasting effects on the development of offspring as well as on their behaviour, physiology and morphology as adults. In some cases, these changes may increase offspring fitness but, in others, they can elevate parental fitness at a cost to the fitness of their offspring. We show that in Kalahari meerkats ( Suricata suricatta), the circulating glucocorticoid (GC) hormones of pregnant females affect the growth and cooperative behaviour of their offspring. We performed a 3-year experiment in wild meerkats to test the hypothesis that GC-mediated maternal effects reduce the potential for offspring to reproduce directly and therefore cause them to exhibit more cooperative behaviour. Daughters (but not sons) born to mothers treated with cortisol during pregnancy grew more slowly early in life and exhibited significantly more of two types of cooperative behaviour (pup rearing and feeding) once they were adults compared to offspring from control mothers. They also had lower measures of GCs as they aged, which could explain the observed increases in cooperative behaviour. Because early life growth is a crucial determinant of fitness in female meerkats, our results indicate that GC-mediated maternal effects may reduce the fitness of offspring, but may elevate parental fitness as a consequence of increasing the cooperative behaviour of their daughters. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Ben Dantzer
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Constance Dubuc
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
| | - Ines Braga Goncalves
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
| | - Dominic L. Cram
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
| | - Nigel C. Bennett
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, 0002 Pretoria, South Africa
| | - Andre Ganswindt
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, 0002 Pretoria, South Africa
- Endocrine Research Laboratory, Department of Anatomy and Physiology, University of Pretoria, Onderstepoort 0110, South Africa
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Chris Duncan
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
| | - David Gaynor
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, 0002 Pretoria, South Africa
| | - Tim H. Clutton-Brock
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
- Kalahari Meerkat Project, Kuruman River Reserve, Northern Cape, South Africa
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, 0002 Pretoria, South Africa
| |
Collapse
|
193
|
Watanabe Y, Abe H, Nakajima K, Ideta-Otsuka M, Igarashi K, Woo GH, Yoshida T, Shibutani M. Aberrant Epigenetic Gene Regulation in GABAergic Interneuron Subpopulations in the Hippocampal Dentate Gyrus of Mouse Offspring Following Developmental Exposure to Hexachlorophene. Toxicol Sci 2019; 163:13-25. [PMID: 29301063 PMCID: PMC5917777 DOI: 10.1093/toxsci/kfx291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Maternal hexachlorophene (HCP) exposure causes transient disruption of hippocampal neurogenesis in mouse offspring. We examined epigenetically hypermethylated and downregulated genes related to this HCP-induced disrupted neurogenesis. Mated female mice were dietary exposed to 0 or 100 ppm HCP from gestational day 6 to postnatal day (PND) 21 on weaning. The hippocampal dentate gyrus of male offspring was subjected to methyl-capture sequencing and real-time reverse transcription-polymerase chain reaction analyses on PND 21. Validation analyses on methylation identified three genes, Dlx4, Dmrt1, and Plcb4, showing promoter-region hypermethylation. Immunohistochemically, DLX4+, DMRT1+, and PLCB4+ cells in the dentate hilus co-expressed GAD67, a γ-aminobutyric acid (GABA)ergic neuron marker. HCP decreased all of three subpopulations as well as GAD67+ cells on PND 21. PLCB4+ cells also co-expressed the metabotropic glutamate receptor, GRM1. HCP also decreased transcript level of synaptic plasticity-related genes in the dentate gyrus and immunoreactive granule cells for synaptic plasticity-related ARC. On PND 77, all immunohistochemical cellular density changes were reversed, whereas the transcript expression of the synaptic plasticity-related genes fluctuated. Thus, HCP-exposed offspring transiently reduced the number of GABAergic interneurons. Among them, subpopulations expressing DLX4, DMRT1, or PLCB4 were transiently reduced in number through an epigenetic mechanism. Considering the role of the Dlx gene family in GABAergic interneuron migration and differentiation, the decreased number of DLX4+ cells may be responsible for reducing those GABAergic interneurons regulating neurogenesis. The effect on granule cell synaptic plasticity was sustained until the adult stage, and reduced GABAergic interneurons active in GRM1–PLCB4 signaling may be responsible for the suppression on weaning.
Collapse
Affiliation(s)
- Yousuke Watanabe
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan.,Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences, Gifu University, Gifu-shi, Gifu 501-1193, Japan
| | - Hajime Abe
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| | - Kota Nakajima
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan.,Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences, Gifu University, Gifu-shi, Gifu 501-1193, Japan
| | - Maky Ideta-Otsuka
- Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Katsuhide Igarashi
- Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Gye-Hyeong Woo
- Laboratory of Histopathology, Department of Clinical Laboratory Science, Semyung University, Jecheon-si, Chungbuk 27136, Republic of Korea
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan.,Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| |
Collapse
|
194
|
Niu X, Wu X, Ying A, Shao B, Li X, Zhang W, Lin C, Lin Y. Maternal high fat diet programs hypothalamic-pituitary-adrenal function in adult rat offspring. Psychoneuroendocrinology 2019; 102:128-138. [PMID: 30544004 DOI: 10.1016/j.psyneuen.2018.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 02/04/2023]
Abstract
Maternal environmental factors such as diet have profound effects on offspring development and later health. The hypothalamic-pituitary-adrenal (HPA) axis is an important stress neuroendocrine system that is subject to programming by early life challenges. The present study was further to investigate whether maternal high fat diet (HFD) exposure during rat pregnancy and lactation can alter the HPA axis activity in adult male offspring. We observed that maternal HFD consumption exerted long-term effects on the basal activity of the HPA axis in adult offspring, with increased mean plasma corticosterone levels that result from elevated steroid pulse frequence and pulse amplitude. More importantly, maternal HFD offspring displayed enhanced corticosterone responses to restraint (1 h) and lipopolysaccharide (25 μg/kg, iv) but not insulin-induced hypoglycemia (0.3U/kg, iv) stress, suggesting a stressor-specific effect of maternal diet on the hyperresponsiveness of the HPA axis to stress. Additionally, maternal HFD exposure markedly attenuated the habituation of HPA responses to repeated restraint stress. These findings demonstrate that perinatal HFD exposure has a potent and long-lasting influence on development of neuroendocrine regulatory mechanisms. Maternal HFD consumption significantly increased basal corticotropin-releasing factor (CRF) mRNA expression in the paraventricular nucleus; nevertheless, similar increments in CRF mRNA levels following restraint were observed between maternal HFD offspring and control rats. Furthermore, the medial and central nuclei of amygdala played a pivotal role in maternal HFD-induced sensitization of the HPA response to psychological and systemic stress, respectively, suggesting that different neural pathways may mediate maternal HFD-induced HPA hyperresponsivity to different types of stressors. Take together, the long-term effects of maternal HFD challenge on the central regulation of the HPA axis, therefore, expose the adult offspring to greater HPA function throughout lifespan, in stressor-specific and region-specific manner.
Collapse
Affiliation(s)
- XiaoTing Niu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoYun Wu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - AnNa Ying
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoFeng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - WanLi Zhang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - ChengCheng Lin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - YuanShao Lin
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
195
|
Russell JA, Brunton PJ. Giving a good start to a new life via maternal brain allostatic adaptations in pregnancy. Front Neuroendocrinol 2019; 53:100739. [PMID: 30802468 DOI: 10.1016/j.yfrne.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/29/2019] [Accepted: 02/21/2019] [Indexed: 12/23/2022]
Abstract
Successful pregnancy requires adjustments to multiple maternal homeostatic mechanisms, governed by the maternal brain to support and enable survival of the growing fetus and placenta. Such adjustments fit the concept of allostasis (stability through change) and have a cost: allostatic load. Allostasis is driven by ovarian, anterior pituitary, placental and feto-placental hormones acting on the maternal brain to promote adaptations that support the pregnancy and protect the fetus. Many women carry an existing allostatic load into pregnancy, from socio-economic circumstances, poor mental health and in 'developed' countries, also from obesity. These pregnancies have poorer outcomes indicating negative interactions (failing allostasis) between pre-pregnancy and pregnancy allostatic loads. Use of animal models, such as adult prenatally stressed female offspring with abnormal neuroendocrine, metabolic and behavioural phenotypes, to probe gene expression changes, and epigenetic mechanisms in the maternal brain in adverse pregnancies are discussed, with the prospect of ameliorating poor pregnancy outcomes.
Collapse
Affiliation(s)
- John A Russell
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; Zhejiang University-University of Edinburgh Institute, Haining, Zhejiang, PR China.
| |
Collapse
|
196
|
Montenegro YHA, de Queiroga Nascimento D, de Assis TO, Santos-Lopes SSD. The epigenetics of the hypothalamic-pituitary-adrenal axis in fetal development. Ann Hum Genet 2019; 83:195-213. [PMID: 30843189 DOI: 10.1111/ahg.12306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/16/2023]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is an important hormonal mechanism of the human body and is extremely programmable during embryonic and fetal development. Analyzing its development in this period is the key to understanding in fact how vulnerabilities of congenital diseases occur and any other changes in the phenotypic and histophysiological aspects of the fetus. The environment in which the mother is exposed during the gestational period can influence this axis. Knowing this, our objective was to analyze in recent research the possible impact of epigenetic programming on the HPA axis and its consequences for fetal development. This review brought together articles from two databases: ScienceDirect and PUBMED researched based on key words such as "epigenetics, HPA axis, cardiovascular disease, and circulatory problems" where it demonstrated full relevance in experimental and scientific settings. A total of 101 articles were selected following the criteria established by the researchers. Thus, it was possible to verify that the development of the HPA axis is directly related to changes that occur in the cardiovascular system, to the cerebral growth and other systems depending on the influence that it receives in the period of fetal formation.
Collapse
|
197
|
Campbell RK, Devick KL, Coull BA, Cowell W, Askowitz T, Goldson B, Wright RO, Wright RJ. Prenatal cortisol modifies the association between maternal trauma history and child cognitive development in a sex-specific manner in an urban pregnancy cohort. Stress 2019; 22:228-235. [PMID: 30767640 PMCID: PMC6476658 DOI: 10.1080/10253890.2018.1553950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/25/2018] [Indexed: 12/16/2022] Open
Abstract
Women's experience of trauma may cause lifelong alterations in physiological stress regulation, which can be transmitted to offspring in utero. We investigated, in a prospective pregnancy cohort, associations among maternal lifetime interpersonal trauma (IPT) history, prenatal cortisol dysregulation, and children's memory domains. Sex-specific effects were also explored. Pregnant women were enrolled from Brigham & Women's Hospital and affiliated clinics near Boston, MA, in 2002-2007. IPT was assessed with the Revised Conflict Tactics Scale, short form. Salivary cortisol was measured at five time points on each of three days in one week at 29.0 ± 5.1 weeks gestation, and morning rise and diurnal slope were calculated. The Wide Range Assessment of Memory & Learning, 2nd Edition was administered at 6.5 ± 1.0 years and scores were generated for general memory and three sub-domains: verbal, visual, and attention/concentration. In total, 258 maternal-child dyads provided memory and IPT and/or cortisol data. IPT was positively associated with verbal memory in boys (β ± SE: 4.6 ± 2.6) and inversely associated with visual memory score in girls (-6.5 ± 3.2). IPT did not predict prenatal cortisol, but prenatal cortisol modified the association between IPT history and child memory in varying coefficient models allowing for non-linear effect modification. The strongest evidence of interaction was for visual memory in boys: IPT history was associated with poorer visual memory only in those with flatter prenatal diurnal slope (interaction p = .005). Maternal lifetime IPT that leads to prenatal HPA dysregulation may have consequences for child memory, more so than either trauma or elevated cortisol alone. Boys may be more vulnerable to effects. Sex- and timing-specific effects require further investigation.
Collapse
Affiliation(s)
- Rebecca K. Campbell
- Department of Pediatrics, Kravis Children’s
Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katrina L. Devick
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, MA, USA
| | - Brent A. Coull
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, MA, USA
| | - Whitney Cowell
- Department of Pediatrics, Kravis Children’s
Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Talia Askowitz
- Department of Pediatrics, Kravis Children’s
Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brandon Goldson
- Department of Pediatrics, Kravis Children’s
Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert O. Wright
- Department of Environmental Medicine and Public Health,
Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J. Wright
- Department of Pediatrics, Kravis Children’s
Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
198
|
Barke TL, Money KM, Du L, Serezani A, Gannon M, Mirnics K, Aronoff DM. Sex modifies placental gene expression in response to metabolic and inflammatory stress. Placenta 2019; 78:1-9. [PMID: 30955704 PMCID: PMC6461364 DOI: 10.1016/j.placenta.2019.02.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/25/2019] [Accepted: 02/18/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Metabolic stress (e.g., gestational diabetes mellitus (GDM) and obesity) and infections are common during pregnancy, impacting fetal development and the health of offspring. Such antenatal stresses can differentially impact male and female offspring. We sought to determine how metabolic stress and maternal immune activation (MIA), either alone or in combination, alters inflammatory gene expression within the placenta and whether the effects exhibited sexual dimorphism. METHODS Female C57BL/6 J mice were fed a normal diet or a high fat diet for 6 weeks prior to mating, with the latter diet inducing a GDM phenotype during pregnancy. Dams within each diet group at gestational day (GD) 12.5 received either an intraperitoneal injection of the viral mimic, polyinosinic:polycytidylic acid (poly(I:C)) or saline. Three hours post injection; placentae were collected and analyzed for changes in the expression of 248 unique immune genes. RESULTS Placental immune gene expression was significantly altered by GDM, MIA and the combination of the two (GDM+MIA). mRNA expression was generally lower in placentae of mice exposed to GDM alone compared with the other experimental groups, while mice exposed to MIA exhibited the highest transcript levels. Notably, fetal/placental sex influenced the responses of many immune genes to both metabolic and inflammatory stress. DISCUSSION GDM and MIA provoke inflammatory responses within the placenta and such effects exhibit sexual dimorphism. The combination of these stressors impacts the placenta differently than either condition alone. These findings may help explain sexual dimorphism observed in adverse pregnancy outcomes in human offspring exposed to similar stressors.
Collapse
Affiliation(s)
- Theresa L Barke
- Graduate Program in Microbiology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Kelli M Money
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Liping Du
- Center for Quantitative Sciences, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ana Serezani
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Maureen Gannon
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - David M Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
199
|
Tanaka T, Nakajima K, Masubuchi Y, Ito Y, Kikuchi S, Ideta-Ohtsuka M, Woo GH, Yoshida T, Igarashi K, Shibutani M. Aberrant epigenetic gene regulation in hippocampal neurogenesis of mouse offspring following maternal exposure to 3,3'-iminodipropionitrile. J Toxicol Sci 2019; 44:93-105. [PMID: 30726815 DOI: 10.2131/jts.44.93] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Maternal exposure to 3,3'-iminodipropionitrile (IDPN) affects hippocampal neurogenesis in mouse offspring, with biphasic disruption, which facilitates neurogenesis during exposure and reduces the broad range of the granule cell lineage population at the adult stage. The present study investigated the epigenetically hypermethylated and downregulated genes related to the IDPN-induced disrupted neurogenesis. Mated female mice were treated with IDPN at 0 or 1200 ppm in drinking water from gestational day 6 to postnatal day (PND) 21 on weaning. The hippocampal dentate gyrus of male offspring on PND 21 was subjected to methyl-capture sequencing and real-time reverse transcription-PCR analyses, followed by validation analyses on DNA methylation. Three genes, Edc4, Kiss1 and Mrpl38, were identified as those showing promoter-region hypermethylation and transcript downregulation, with Mrpl38 sustaining the changes through PND 77. Immunohistochemically, MRPL38, a mitochondrial ribosomal protein, revealed an irreversible decrease in the number of immunoreactive interneurons in the dentate gyrus hilar region, suggesting a causal relationship with the long-lasting effect on neurogenesis by the impaired migration due to mitochondrial dysfunction of interneurons, which regulate the differentiation and survival of granule cell lineages. Downregulation of Edc4 may also be responsible for decreased neurogenesis on PND 77 owing to a mechanism involving interleukin-6 downregulation via processing body dysfunction. Downregulation of Kiss1 may be responsible for the facilitation of neurogenesis during IDPN-exposure due to decreased glutamatergic neurotransmission and also for suppressed neurogenesis on PND 77 due to decreased expression of immediate-early genes, which play a crucial role in the maintenance of cell differentiation or plasticity.
Collapse
Affiliation(s)
- Takeshi Tanaka
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology
| | - Kota Nakajima
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology.,Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences, Gifu University
| | - Yasunori Masubuchi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology.,Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences, Gifu University
| | - Yuko Ito
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology.,Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences, Gifu University
| | - Satomi Kikuchi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology.,Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology
| | - Maky Ideta-Ohtsuka
- Laboratory of Biofunctional Science, School of Pharmacy and Pharmaceutical Sciences, Hoshi University
| | - Gye-Hyeong Woo
- Laboratory of Histopathology, Department of Clinical Laboratory Science, Semyung University, Korea
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology
| | - Katsuhide Igarashi
- Laboratory of Biofunctional Science, School of Pharmacy and Pharmaceutical Sciences, Hoshi University
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology.,Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology.,Institute of Global Innovation Research, Tokyo University of Agriculture and Technology
| |
Collapse
|
200
|
Genome-wide epigenetic signatures of childhood adversity in early life: Opportunities and challenges. J Dev Orig Health Dis 2019; 10:65-72. [PMID: 30744719 DOI: 10.1017/s2040174418000843] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Maternal adversity and fetal glucocorticoid exposure has long-term effects on cardiovascular, metabolic and behavioral systems in offspring that can persist throughout the lifespan. These data, along with other environmental exposure data, implicate epigenetic modifications as potential mechanisms for long-term effects of maternal exposures on adverse health outcomes in offspring. Advances in microarray, sequencing and bioinformatic approaches have enabled recent studies to examine the genome-wide epigenetic response to maternal adversity. Studies of maternal exposures to xenobiotics such as arsenic and smoking have been performed at birth to examine fetal epigenomic signatures in cord blood relating to adult health outcomes. However, there have been no epigenomic studies examining these effects in animal models. On the other hand, to date, only a few studies of the effects of maternal psychosocial stress have been performed in human infants, and the majority of animal studies have examined epigenomic outcomes in adulthood. In terms of maternal exposure to excess glucocorticoids by synthetic glucocorticoid treatment, there has been no epigenetic study performed in humans and only a few studies undertaken in animal models. This review emphasizes the importance of examining biomarkers of exposure to adversity throughout development to identify individuals at risk and to target interventions. Thus, research performed at birth will be reviewed. In addition, potential subject characteristics associated with epigenetic modifications, technical considerations, the selection of target tissues and combining human studies with animal models will be discussed in relation to the design of experiments in this field of study.
Collapse
|