1
|
Maurer SV, Hing BWQ, Lussier S, Radhakrishna S, Davis JLB, Abbott PW, Michaelson JJ, Stevens HE. Prenatal stress alters mouse offspring dorsal striatal development and placental function in sex-specific ways. J Psychiatr Res 2025; 182:149-160. [PMID: 39809011 PMCID: PMC11959308 DOI: 10.1016/j.jpsychires.2024.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Prenatal stress is a risk factor for neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD). However, how early stress modification of brain development contributes to this pathophysiology is poorly understood. Ventral forebrain regions such as dorsal striatum are of particular interest: dorsal striatum modulates movement and cognition, is altered in NDDs, and has a primarily GABAergic population. Here, we examine effects of prenatal stress on adult movement, cognition, and dorsal striatum neurobiology in mice using striatal-dependent behavioral assays, immunohistochemistry, embryonic ventral forebrain transcriptomics, and placental transcriptomics. We found prenatal stress affected adult procedural, habit, and reversal learning in sex-specific ways. Stress also increased adult dorsal striatal GABAergic neurons - an effect largely driven by males. We sought to examine the developmental origins of these adult brain changes. We found similar sex-specific dorsal striatal cellular changes in earlier points of development. The dorsal striatum primordium--embryonic ventral forebrain-showed that prenatal stress increased DNA replication and cell cycle pathways in male but not female transcriptomics and cellular biology. Unique signatures may have arisen from male-female placental differences. Stress-induced placental transcriptomics showed upregulated morphogenesis pathways in males while females downregulated morphogenic, hormonal, and cellular response pathways. Our findings suggest that prenatal stress could affect placenta function and also alter the GABAergic population of dorsal striatum differentially between the sexes.
Collapse
Affiliation(s)
- Sara V Maurer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Benjamin W Q Hing
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Stephanie Lussier
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SL is now with Moderna, USA
| | - Sreya Radhakrishna
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; SR is now at Albert Einstein College of Medicine, USA
| | - Jada L B Davis
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Parker W Abbott
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52246, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
2
|
Bodin R, Seewooruttun C, Corona A, Delanaud S, Pelletier A, Villégier AS. Sex-dependent impact of perinatal 5G electromagnetic field exposure in the adolescent rat behavior. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:113704-113717. [PMID: 37851267 DOI: 10.1007/s11356-023-30256-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023]
Abstract
The fifth generation (5G) network is currently being worldwide spread out, raising questions about the potential impact of this new technology, particularly on immature organisms. The current study aimed to investigate the effects of daily 5G electromagnetic field (EMF) perinatal exposure on the neurodevelopment of rats. The exposure level was set to the limit of whole-body public exposure defined by the International Commission on Non-Ionizing Radiation Protection. The mother rat specific absorption rate (SAR) was 0.07 W/kg for 22 h/day at 3500 MHz continuous wave from gestational day (GD) 8 to post-natal day (PND) 21. Clinical observations were performed on weight, length, sex ratio, number of pups per litter, and number of stillborn in sham and EMF-exposed groups (n = 7). The age of pinna ear detachment, incisor eruption, and eye opening were recorded. Behavior was assessed on righting, gripping, and negative geotaxis reflexes at PND 3 or 7 and on stereotyped and horizontal movements in the open field at PND 43. Our results indicated that both male and female pups showed delayed incisor eruption in the EMF-exposed group compared to the sham group (+ 1 day). Regarding activity in the open field, adolescent females showed less stereotyped movements (- 70%), while adolescent males showed more stereotyped movements (+ 50%) compared to the sham-exposed adolescent rats. Thus, the present study suggested that perinatal exposure to 5G at SAR level below reglementary threshold led to perturbations in the descendants seen in juveniles and adolescents.
Collapse
Affiliation(s)
- Raphaël Bodin
- PERITOX Laboratory (UMR_I 01), INERIS, MIV/TEAM, Verneuil-en-Halatte, France
| | | | - Aurélie Corona
- University of Picardie Jules Verne, CURS, Amiens, France
| | | | | | | |
Collapse
|
3
|
Soti M, Ranjbar H, Kohlmeier KA, Shabani M. Sex differences in the vulnerability of the hippocampus to prenatal stress. Dev Psychobiol 2022; 64:e22305. [PMID: 36282753 DOI: 10.1002/dev.22305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 05/28/2022] [Indexed: 01/27/2023]
Abstract
Distressing events during pregnancy that engage activity of the body's endocrine stress response have been linked with later life cognitive deficits in offspring and associated with developmental changes in cognitive-controlling neural regions. Interestingly, prenatal stress (PS)-induced alterations have shown some sex specificity. Here, we review the literature of animal studies examining sex-specific effect of physical PS on the function and structure of the hippocampus as hippocampal impairments likely underlie PS-associated deficits in learning and memory. Furthermore, the connectivity between the hypothalamic-pituitary-adrenal (HPA) axis and the hippocampus as well as the heavy presence of glucocorticoid receptors (GRs) in the hippocampus suggests this structure plays an important role in modulation of activity within stress circuitry in a sex-specific pattern. We hope that better understanding of sex-specific, PS-related hippocampal impairment will assist in uncovering the molecular mechanisms behind sex-based risk factors in PS populations across development, and perhaps contribute to greater precision in management of cognitive disturbances in this vulnerable population.
Collapse
Affiliation(s)
- Monavareh Soti
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Hoda Ranjbar
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
4
|
Groenewold NA, Wedderburn CJ, Pellowski JA, Fouché JP, Michalak L, Roos A, Woods RP, Narr KL, Zar HJ, Donald KA, Stein DJ. Subcortical brain volumes in young infants exposed to antenatal maternal depression: Findings from a South African birth cohort. Neuroimage Clin 2022; 36:103206. [PMID: 36162238 PMCID: PMC9668606 DOI: 10.1016/j.nicl.2022.103206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Several studies have reported enlarged amygdala and smaller hippocampus volumes in children and adolescents exposed to maternal depression. It is unclear whether similar volumetric differences are detectable in the infants' first weeks of life, following exposure in utero. We investigated subcortical volumes in 2-to-6 week old infants exposed to antenatal maternal depression (AMD) from a South African birth cohort. METHODS AMD was measured with the Beck Depression Inventory 2nd edition (BDI-II) at 28-32 weeks gestation. T2-weighted structural images were acquired during natural sleep on a 3T Siemens Allegra scanner. Subcortical regions were segmented based on the University of North Carolina neonatal brain atlas. Volumetric estimates were compared between AMD-exposed (BDI-II ⩾ 20) and unexposed (BDI-II < 14) infants, adjusted for age, sex and total intracranial volume using analysis of covariance. RESULTS Larger volumes were observed in AMD-exposed (N = 49) compared to unexposed infants (N = 75) for the right amygdala (1.93% difference, p = 0.039) and bilateral caudate nucleus (left: 5.79% difference, p = 0.001; right: 6.09% difference, p < 0.001). A significant AMD-by-sex interaction was found for the hippocampus (left: F(1,118) = 4.80, p = 0.030; right: F(1,118) = 5.16, p = 0.025), reflecting greater volume in AMD-exposed females (left: 5.09% difference, p = 0.001, right: 3.54% difference, p = 0.010), but not males. CONCLUSIONS Volumetric differences in subcortical regions can be detected in AMD-exposed infants soon after birth, suggesting structural changes may occur in utero. Female infants might exhibit volumetric changes that are not observed in male infants. The potential mechanisms underlying these early volumetric differences, and their significance for long-term child mental health, require further investigation.
Collapse
Affiliation(s)
- Nynke A Groenewold
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SA-MRC) Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa; Department of Psychiatry & Mental Health, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
| | - Catherine J Wedderburn
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jennifer A Pellowski
- Department of Behavioral and Social Sciences and International Health Institute, Brown University School of Public Health, Providence, RI, USA; Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Jean-Paul Fouché
- Department of Psychiatry & Mental Health, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Liza Michalak
- Department of Psychiatry & Mental Health, University of Cape Town, Cape Town, South Africa
| | - Annerine Roos
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa; SA-MRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Roger P Woods
- Departments of Neurology, Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, USA
| | - Katherine L Narr
- Departments of Neurology, Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, USA
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; South African Medical Research Council (SA-MRC) Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Department of Psychiatry & Mental Health, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa; SA-MRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
6
|
Yao D, Mu Y, Lu Y, Li L, Shao S, Zhou J, Li J, Chen S, Zhang D, Zhang Y, Zhu Z, Li H. Hippocampal AMPA receptors mediate the impairment of spatial learning and memory in prenatally stressed offspring rats. J Psychiatr Res 2022; 151:17-24. [PMID: 35427874 DOI: 10.1016/j.jpsychires.2022.03.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/15/2022] [Accepted: 03/21/2022] [Indexed: 11/18/2022]
Abstract
Numerous studies have shown that prenatal stress (PS) induces learning and memory deficits in offspring, yet the specific mechanisms and effective interventions remain limited. Chewing has been known as one of the active coping strategies to suppress stress, but its effects during PS on learning and memory are unknown. The purpose of this study was to investigate the role of hippocampal AMPA receptors in the adverse effects of PS on spatial learning and memory, and whether chewing during PS could prevent these effects in prenatally stressed adult offspring rats. Prenatal restraint stress with or without chewing to dams during the day 11-20 of pregnancy was used to analyze the impact of different treatments for offspring. The spatial learning and memory were tested by the Morris water maze. The mRNA and protein expression of AMPA receptors in the hippocampus were measured by qRT-PCR and Western blot, respectively. The methylation of AMPA receptors was detected by bisulfite sequencing PCR. Our results revealed that PS impaired spatial learning acquisition and memory retrieval in adult offspring rats, but chewing could relieve this effect. Hippocampal GluA1-4 expression was significantly reduced in prenatally stressed offspring, while there were no changes in the methylation level of GluA2 and GluA4 promoters. Moreover, chewing increased PS-induced suppression of AMPA receptors in the hippocampus. In short, hippocampal AMPA receptors mediate the impairment of spatial learning and memory in prenatally stressed offspring, whereas chewing during PS could ameliorate PS-induced memory deficits.
Collapse
Affiliation(s)
- Dan Yao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yingjun Mu
- Central Laboratory, Heze Medical College, 1750 University Road, Heze, Shandong, 274009, China
| | - Yong Lu
- Central Laboratory, Heze Medical College, 1750 University Road, Heze, Shandong, 274009, China
| | - Li Li
- Central Laboratory, Heze Medical College, 1750 University Road, Heze, Shandong, 274009, China
| | - Shuya Shao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jiahao Zhou
- Maternal and Infant Health Research Institute, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Jing Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Shengquan Chen
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Dan Zhang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yifan Zhang
- Central Laboratory, Heze Medical College, 1750 University Road, Heze, Shandong, 274009, China
| | - Zhongliang Zhu
- Maternal and Infant Health Research Institute, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
7
|
Kocamaz D, Franzke C, Gröger N, Braun K, Bock J. Early Life Stress-Induced Epigenetic Programming of Hippocampal NPY-Y2 Receptor Gene Expression Changes in Response to Adult Stress. Front Cell Neurosci 2022; 16:936979. [PMID: 35846564 PMCID: PMC9283903 DOI: 10.3389/fncel.2022.936979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Early Life Stress (ELS) can critically influence brain development and future stress responses and thus represents an important risk factor for mental health and disease. Neuropeptide Y (NPY) is discussed to be a key mediator of resilient vs. vulnerable adaptations and specifically, the NPY-Y2 receptor (Y2R) may be involved in the pathophysiology of depression due to its negative regulation of NPY-release. The present study addressed the hypotheses that ELS and adult stress (AS) affect the expression of hippocampal Y2R and that exposure to ELS induces an epigenetically mediated programming effect towards a consecutive stress exposure in adulthood. The specific aims were to investigate if (i) ELS or AS as single stressors induce changes in Y2 receptor gene expression in the hippocampus, (ii) the predicted Y2R changes are epigenetically mediated via promoter-specific DNA-methylation, (iii) the ELS-induced epigenetic changes exert a programming effect on Y2R gene expression changes in response to AS, and finally (iv) if the predicted alterations are sex-specific. Animals were assigned to the following experimental groups: (1) non-stressed controls (CON), (2) only ELS exposure (ELS), (3) only adult stress exposure (CON+AS), and (4) exposure to ELS followed by AS (ELS+AS). Using repeated maternal separation in mice as an ELS and swim stress as an AS we found that both stressors affected Y2R gene expression in the hippocampus of male mice but not in females. Specifically, upregulated expression was found in the CON+AS group. In addition, exposure to both stressors ELS+AS significantly reduced Y2R gene expression when compared to CON+AS. The changes in Y2R expression were paralleled by altered DNA-methylation patterns at the Y2R promoter, specifically, a decrease in mean DNA-methylation in the CON+AS males compared to the non-AS exposed groups and an increase in the ELS+AS males compared to the CON+AS males. Also, a strong negative correlation of mean DNA-methylation with Y2R expression was found. Detailed CpG-site-specific analysis of DNA-methylation revealed that ELS induced increased DNA-methylation only at specific CpG-sites within the Y2R promoter. It is tempting to speculate that these ELS-induced CpG-site-specific changes represent a “buffering” programming effect against elevations of Y2R expression induced by AS.
Collapse
Affiliation(s)
- Derya Kocamaz
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Caroline Franzke
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Nicole Gröger
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Katharina Braun
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Jörg Bock
- Center for Behavioral Brain Sciences, Magdeburg, Germany
- PG “Epigenetics and Structural Plasticity,” Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- *Correspondence: Jörg Bock,
| |
Collapse
|
8
|
Juvenile handling rescues autism-related effects of prenatal exposure to valproic acid. Sci Rep 2022; 12:7174. [PMID: 35504947 PMCID: PMC9065111 DOI: 10.1038/s41598-022-11269-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/13/2022] [Indexed: 11/12/2022] Open
Abstract
Environmental factors acting on young animals affect neurodevelopmental trajectories and impact adult brain function and behavior. Psychiatric disorders may be caused or worsen by environmental factors, but early interventions can improve performance. Understanding the possible mechanisms acting upon the developing brain could help identify etiological factors of psychiatric disorders and enable advancement of effective therapies. Research has focused on the long-lasting effects of environmental factors acting during the perinatal period, therefore little is known about the impact of these factors at later ages when neurodevelopmental pathologies such as autism spectrum disorder (ASD) are usually diagnosed. Here we show that handling mice during the juvenile period can rescue a range of behavioral and cellular effects of prenatal valproic acid (VPA) exposure. VPA-exposed animals show reduced sociability and increased repetitive behaviors, along with other autism-related endophenotypes such as increased immobility in the forced swim test and increased neuronal activity in the piriform cortex (Pir). Our results demonstrate that briefly handling mice every other day between postnatal days 22 and 34 can largely rescue these phenotypes. This effect can also be observed when animals are analyzed across tests using an “autism” factor, which also discriminates between animals with high and low Pir neuron activity. Thus, we identified a juvenile developmental window when environmental factors can determine adult autism-related behavior. In addition, our results have broader implications on behavioral neuroscience, as they highlight the importance of adequate experimental design and control of behavioral experiments involving treating or testing young animals.
Collapse
|
9
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
10
|
Gaspar R, Soares-Cunha C, Domingues AV, Coimbra B, Baptista FI, Pinto L, Ambrósio AF, Rodrigues AJ, Gomes CA. The Duration of Stress Determines Sex Specificities in the Vulnerability to Depression and in the Morphologic Remodeling of Neurons and Microglia. Front Behav Neurosci 2022; 16:834821. [PMID: 35330844 PMCID: PMC8940280 DOI: 10.3389/fnbeh.2022.834821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Stress exposure has been shown to induce a variety of molecular and functional alterations associated with anxiety and depression. Some studies suggest that microglia, the immune cells of the brain, play a significant role in determining neuronal and behavioral responses to chronic stress and also contribute to the development of stress-related psychopathologies. However, little is known about the impact of the duration of stress exposure upon microglia and neurons morphology, particularly considering sex differences. This issue deserves particular investigation, considering that the process of morphologic remodeling of neurons and microglia is usually accompanied by functional changes with behavioral expression. Here, we examine the effects of short and long unpredictable chronic mild stress (uCMS) protocols on behavior, evaluating in parallel microglia and neurons morphology in the dorsal hippocampus (dHIP) and in the nucleus accumbens (NAc), two brain regions involved in the etiology of depression. We report that long-term uCMS induced more behavioral alterations in males, which present anxiety and depression-like phenotypes (anhedonia and helplessness behavior), while females only display anxiety-like behavior. After short-term uCMS, both sexes presented anxiety-like behavior. Microglia cells undergo a process of morphologic adaptation to short-term uCMS, dependent on sex, in the NAc: we observed a hypertrophy in males and an atrophy in females, transient effects that do not persist after long-term uCMS. In the dHIP, the morphologic adaptation of microglia is only observed in females (hypertrophy) and after the protocol of long uCMS. Interestingly, males are more vulnerable to neuronal morphological alterations in a region-specific manner: dendritic atrophy in granule neurons of the dHIP and hypertrophy in the medium spiny neurons of the NAc, both after short- or long-term uCMS. The morphology of neurons in these brain regions were not affected in females. These findings raise the possibility that, by differentially affecting neurons and microglia in dHIP and NAc, chronic stress may contribute for differences in the clinical presentation of stress-related disorders under the control of sex-specific mechanisms.
Collapse
Affiliation(s)
- Rita Gaspar
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Verónica Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bárbara Coimbra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa I. Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António F. Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
- *Correspondence: Ana João Rodrigues,
| | - Catarina A. Gomes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Catarina A. Gomes,
| |
Collapse
|
11
|
Sikes-Keilp C, Rubinow DR. In search of sex-related mediators of affective illness. Biol Sex Differ 2021; 12:55. [PMID: 34663459 PMCID: PMC8524875 DOI: 10.1186/s13293-021-00400-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022] Open
Abstract
Sex differences in the rates of affective disorders have been recognized for decades. Studies of physiologic sex-related differences in animals and humans, however, have generally yielded little in terms of explaining these differences. Furthermore, the significance of these findings is difficult to interpret given the dynamic, integrative, and highly context-dependent nature of human physiology. In this article, we provide an overview of the current literature on sex differences as they relate to mood disorders, organizing existing findings into five levels at which sex differences conceivably influence physiology relevant to affective states. These levels include the following: brain structure, network connectivity, signal transduction, transcription/translation, and epigenesis. We then evaluate the importance and limitations of this body of work, as well as offer perspectives on the future of research into sex differences. In creating this overview, we attempt to bring perspective to a body of research that is complex, poorly synthesized, and far from complete, as well as provide a theoretical framework for thinking about the role that sex differences ultimately play in affective regulation. Despite the overall gaps regarding both the underlying pathogenesis of affective illness and the role of sex-related factors in the development of affective disorders, it is evident that sex should be considered as an important contributor to alterations in neural function giving rise to susceptibility to and expression of depression.
Collapse
Affiliation(s)
| | - David R Rubinow
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Thomason ME, Hect JL, Waller R, Curtin P. Interactive relations between maternal prenatal stress, fetal brain connectivity, and gestational age at delivery. Neuropsychopharmacology 2021; 46:1839-1847. [PMID: 34188185 PMCID: PMC8357800 DOI: 10.1038/s41386-021-01066-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Studies reporting significant associations between maternal prenatal stress and child outcomes are frequently confounded by correlates of prenatal stress that influence the postnatal rearing environment. The major objective of this study is to identify whether maternal prenatal stress is associated with variation in human brain functional connectivity prior to birth. We utilized fetal fMRI in 118 fetuses [48 female; mean age 32.9 weeks (SD = 3.87)] to evaluate this association and further addressed whether fetal neural differences were related to maternal health behaviors, social support, or birth outcomes. Community detection was used to empirically define networks and enrichment was used to isolate differential within- or between-network connectivity effects. Significance for χ2 enrichment was determined by randomly permuting the subject pairing of fetal brain connectivity and maternal stress values 10,000 times. Mixtures modelling was used to test whether fetal neural differences were related to maternal health behaviors, social support, or birth outcomes. Increased maternal prenatal negative affect/stress was associated with alterations in fetal frontoparietal, striatal, and temporoparietal connectivity (β = 0.82, p < 0.001). Follow-up analysis demonstrated that these associations were stronger in women with better health behaviors, more positive interpersonal support, and lower overall stress (β = 0.16, p = 0.02). Additionally, magnitude of stress-related differences in neural connectivity was marginally correlated with younger gestational age at delivery (β = -0.18, p = 0.05). This is the first evidence that negative affect/stress during pregnancy is reflected in functional network differences in the human brain in utero, and also provides information about how positive interpersonal and health behaviors could mitigate prenatal brain programming.
Collapse
Affiliation(s)
- Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA.
- Department of Population Health, New York University Medical Center, New York, NY, USA.
- Neuroscience Institute, NYU Langone Health, New York, NY, USA.
| | - Jasmine L Hect
- Medical Scientist Training Program, University of Pittsburgh & Carnegie Mellon University, Pittsburgh, PA, USA
| | - Rebecca Waller
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Curtin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Luft C, Haute GV, Wearick-Silva LE, Antunes KH, da Costa MS, de Oliveira JR, Donadio MVF. Prenatal stress and KCl-induced depolarization modulate cell death, hypothalamic-pituitary-adrenal axis genes, oxidative and inflammatory response in primary cortical neurons. Neurochem Int 2021; 147:105053. [PMID: 33961947 DOI: 10.1016/j.neuint.2021.105053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/01/2021] [Accepted: 04/24/2021] [Indexed: 12/27/2022]
Abstract
Maternal stress has been described as an important component in the offspring's cerebral development, altering the susceptibility to diseases in later life. Moreover, the postnatal period is essential for the development and integration of several peripheral and central systems related to the control of homeostasis. Thus, this study aimed to evaluate the effects of prenatal stress on the activation of cortical neurons, by performing experiments both under basal conditions and after KCl-induced depolarization. Female mice were divided in two groups: control and prenatal restraint stress. Cortical neurons from the offspring were obtained at gestational day 18. The effects of prenatal stress and KCl stimulations on cellular mortality, autophagy, gene expression, oxidative stress, and inflammation were evaluated. We found that neurons from PNS mice have decreased necrosis and autophagy after depolarization. Moreover, prenatal stress modulated the HPA axis, as observed by the increased GR and decreased 5HTr1 mRNA expression. The BDNF is an important factor for neuronal function and results demonstrated that KCl-induced depolarization increased the gene expression of BDNF I, BDNF IV, and TRκB. Furthermore, prenatal stress and KCl treatment induced significant alterations in oxidative and inflammatory markers. In conclusion, prenatal stress and stimulation with KCl may influence several markers related to neurodevelopment in cortical neurons from neonate mice, supporting the well-known long-term effects of maternal stress.
Collapse
Affiliation(s)
- Carolina Luft
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Gabriela Viegas Haute
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luís Eduardo Wearick-Silva
- Exercise, Behavior and Cognition Research Group, Psychology Department, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana Severo da Costa
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| |
Collapse
|
14
|
Abramova O, Ushakova V, Zorkina Y, Zubkov E, Storozheva Z, Morozova A, Chekhonin V. The Behavior and Postnatal Development in Infant and Juvenile Rats After Ultrasound-Induced Chronic Prenatal Stress. Front Physiol 2021; 12:659366. [PMID: 33935805 PMCID: PMC8082110 DOI: 10.3389/fphys.2021.659366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/23/2021] [Indexed: 01/30/2023] Open
Abstract
Fetal development is susceptible to environmental factors. One such factor is exposure to stress during pregnancy. The present study aimed to investigate the effects of chronic prenatal stress (PS) on the development and behavior of rat offspring during infancy and juvenile ages. Existing approaches to modeling prenatal stress on animals do not correlate with the main type of stress in pregnant women, namely psychological stress. We used a new stress paradigm in the experiment, namely, stress induced by exposure to variable frequency ultrasound (US), which acted on pregnant Wistar rats on gestational days 1–21. This type of stress in rodents can be comparable to psychological stress in humans. We assessed physical development, reflex maturation, motor ability development, anxious behavior, response to social novelty, and social play behavior in male and female offspring. Additionally, we investigated maternal behavior and the effect of neonatal handling (NH) on behavior. Prenatal stress did not affect postnatal developmental characteristics in rat pups, but prenatally stressed rats had higher body weight in early and adult age than controls. Prenatal exposure to a stressor increased anxiety in the open-field test (OF), changed social preferences in the social novelty test (SN), and impaired social play behavior in males. Neonatal handling reduced anxiety and restored social behavior, but evoked hyperactive behavior in rat pups. Maternal behavior did not change. Our study demonstrated for the first time that exposure to variable frequency ultrasound during pregnancy influences offspring development and impairs behavior, correlating with the effects of other types of stress during pregnancy in rodents. This supports the idea of using this exposure to model prenatal stress.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.,Mental-health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Moscow, Russia
| | - Valeria Ushakova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.,Mental-health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Moscow, Russia.,Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.,Mental-health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Moscow, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Zinaida Storozheva
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Anna Morozova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.,Mental-health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.,Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
15
|
Brown T, McElroy T, Simmons P, Walters H, Ntagwabira F, Wang J, Byrum SD, Allen AR. Cognitive impairment resulting from treatment with docetaxel, doxorubicin, and cyclophosphamide. Brain Res 2021; 1760:147397. [PMID: 33705788 DOI: 10.1016/j.brainres.2021.147397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 01/08/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer among women and it is estimated that about 30% of newly diagnosed cancers in women will be breast cancers. While advancements in treating breast cancer have led to an average 5-year survival rate of 90%, many survivors experience cognitive impairments as a result of chemotherapy treatment. Doxorubicin, cyclophosphamide, and docetaxel (TAC) are commonly administered as breast cancer treatments; however, there are few studies that have tested the cognitive effects of TAC. In the current study, 12-week-old female C57BL/6 mice received 4 weekly intraperitoneal injections of either saline or a combination therapy of doxorubicin and cyclophosphamide followed by 4 weekly docetaxel injections. Four weeks after the last injection, mice were tested for hippocampus-dependent cognitive performance in the Y-maze and the Morris water maze. During Y-maze testing, mice exposed to TAC exhibited impairment. During the water maze assessment, all animals were able to locate the visible and hidden platform locations. However, mice that received the TAC presented with a significant impairment in spatial memory retention on the probe trial days. TAC treatment significantly decreases the dendritic complexity of arborization in the dentate gyrus region of the hippocampus. In addition, comparative proteomic analysis revealed downregulation of proteins within key metabolic and signaling pathways associated with cognitive dysfunction, such as oxidative phosphorylation, ephrin signaling, and calcium signaling.
Collapse
Affiliation(s)
- Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Huddoy Walters
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Fabio Ntagwabira
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Arkansas Children's Research Institute, Little Rock, AR 72202, United States
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
16
|
Fitzgerald E, Parent C, Kee MZL, Meaney MJ. Maternal Distress and Offspring Neurodevelopment: Challenges and Opportunities for Pre-clinical Research Models. Front Hum Neurosci 2021; 15:635304. [PMID: 33643013 PMCID: PMC7907173 DOI: 10.3389/fnhum.2021.635304] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Pre-natal exposure to acute maternal trauma or chronic maternal distress can confer increased risk for psychiatric disorders in later life. Acute maternal trauma is the result of unforeseen environmental or personal catastrophes, while chronic maternal distress is associated with anxiety or depression. Animal studies investigating the effects of pre-natal stress have largely used brief stress exposures during pregnancy to identify critical periods of fetal vulnerability, a paradigm which holds face validity to acute maternal trauma in humans. While understanding these effects is undoubtably important, the literature suggests maternal stress in humans is typically chronic and persistent from pre-conception through gestation. In this review, we provide evidence to this effect and suggest a realignment of current animal models to recapitulate this chronicity. We also consider candidate mediators, moderators and mechanisms of maternal distress, and suggest a wider breadth of research is needed, along with the incorporation of advanced -omics technologies, in order to understand the neurodevelopmental etiology of psychiatric risk.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Carine Parent
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Michelle Z. L. Kee
- Translational Neuroscience Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Michael J. Meaney
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
- Translational Neuroscience Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Becker M, Pinhasov A, Ornoy A. Animal Models of Depression: What Can They Teach Us about the Human Disease? Diagnostics (Basel) 2021; 11:123. [PMID: 33466814 PMCID: PMC7830961 DOI: 10.3390/diagnostics11010123] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Depression is apparently the most common psychiatric disease among the mood disorders affecting about 10% of the adult population. The etiology and pathogenesis of depression are still poorly understood. Hence, as for most human diseases, animal models can help us understand the pathogenesis of depression and, more importantly, may facilitate the search for therapy. In this review we first describe the more common tests used for the evaluation of depressive-like symptoms in rodents. Then we describe different models of depression and discuss their strengths and weaknesses. These models can be divided into several categories: genetic models, models induced by mental acute and chronic stressful situations caused by environmental manipulations (i.e., learned helplessness in rats/mice), models induced by changes in brain neuro-transmitters or by specific brain injuries and models induced by pharmacological tools. In spite of the fact that none of the models completely resembles human depression, most animal models are relevant since they mimic many of the features observed in the human situation and may serve as a powerful tool for the study of the etiology, pathogenesis and treatment of depression, especially since only few patients respond to acute treatment. Relevance increases by the fact that human depression also has different facets and many possible etiologies and therapies.
Collapse
Affiliation(s)
- Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Albert Pinhasov
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| |
Collapse
|
18
|
Shang Y, Chen R, Li F, Zhang H, Wang H, Zhang T. Prenatal stress impairs memory function in the early development of male-offspring associated with the gaba function. Physiol Behav 2021; 228:113184. [DOI: 10.1016/j.physbeh.2020.113184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/15/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022]
|
19
|
Braun K, Bock J, Wainstock T, Matas E, Gaisler-Salomon I, Fegert J, Ziegenhain U, Segal M. Experience-induced transgenerational (re-)programming of neuronal structure and functions: Impact of stress prior and during pregnancy. Neurosci Biobehav Rev 2020; 117:281-296. [DOI: 10.1016/j.neubiorev.2017.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
|
20
|
Effects of Maternal Chewing on Prenatal Stress-Induced Cognitive Impairments in the Offspring via Multiple Molecular Pathways. Int J Mol Sci 2020; 21:ijms21165627. [PMID: 32781547 PMCID: PMC7460630 DOI: 10.3390/ijms21165627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
We aimed to investigate the effects of maternal chewing on prenatal stress-induced cognitive impairments in the offspring and to explore the molecular pathways of maternal chewing in a mice model. Maternal chewing ameliorated spatial learning impairments in the offspring in a Morris water maze test. Immunohistochemistry and Western blot findings revealed that maternal chewing alleviated hippocampal neurogenesis impairment and increased the expression of hippocampal brain-derived neurotrophic factor in the offspring. In addition, maternal chewing increased the expression of glucocorticoid receptor (GR) and 11β-hydroxysteroid dehydrogenase isozyme 2 (11β-HSD2) and decreased the expression of 11β-HSD1 in the placenta, thereby attenuating the increase of glucocorticoid in the offspring. Furthermore, maternal chewing increased the expression of 11β-HSD2, FK506-binding protein 51 (FKBP51) and FKBP52 and decreased the expression of 11β-HSD1, thereby increasing hippocampal nuclear GR level. In addition, maternal chewing attenuated the increase in expression of DNMT1 and DNMT3a and the decrease in expression of histone H3 methylation at lysine 4, 9, 27 and histone H3 acetylation at lysine 9 induced by prenatal stress in the offspring. Our findings suggest that maternal chewing could ameliorate prenatal stress-induced cognitive impairments in the offspring at least in part by protecting placenta barrier function, alleviating hippocampal nuclear GR transport impairment and increasing the hippocampal brain-derived neurotrophic factor (BDNF) level.
Collapse
|
21
|
Ayash S, Schmitt U, Lyons DM, Müller MB. Stress inoculation in mice induces global resilience. Transl Psychiatry 2020; 10:200. [PMID: 32561821 PMCID: PMC7305209 DOI: 10.1038/s41398-020-00889-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Each year, more than half a billion people in the world are affected by stress-related health disorders. Consequently, there is an urgent need for new insights to guide interventions designed to increase stress resilience. Studies of humans and various animals have uncovered the process of stress inoculation, in which exposure to mild stressors enhances subsequent stress resilience. Here, we investigate whether stress inoculation-induced resilience in mice consistently occurs across a multiplicity of different stress contexts (tests). C57BL/6 J adult male mice were randomised either to stress inoculation training (n = 36) or to a non-inoculated, but handled control condition (n = 36). Thereafter, indications of coping and resilience were assessed during (i) acute social defeat in a context similar to that used for stress inoculation training, and (ii) fear conditioning and learned extinction in a novel context. Stress inoculation effects were also assessed during (iii) tail-suspension and (iv) open-field tests that each represent milder stressors. Stress-inoculated mice showed more active defence behaviour during acute social defeat, higher sociability before and after defeat, and greater indications of learned extinction of conditioned fear compared to non-inoculated control mice. Stress-inoculated mice also responded with diminished tail-suspension immobility and open-field defecation. Results suggest that stress inoculation protects against various stressors that differ in quality and relative intensity. Stress inoculation research in mice may serve as the basis for mechanistic studies of global resilience in humans.
Collapse
Affiliation(s)
- Sarah Ayash
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| | - Ulrich Schmitt
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| | - David M Lyons
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Marianne B Müller
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany.
- Leibniz Institute for Resilience Research, Mainz, Germany.
| |
Collapse
|
22
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
23
|
Baratta AM, Kanyuch NR, Cole CA, Valafar H, Deslauriers J, Pocivavsek A. Acute sleep deprivation during pregnancy in rats: Rapid elevation of placental and fetal inflammation and kynurenic acid. Neurobiol Stress 2019; 12:100204. [PMID: 32258253 PMCID: PMC7109515 DOI: 10.1016/j.ynstr.2019.100204] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/27/2019] [Accepted: 12/11/2019] [Indexed: 01/19/2023] Open
Abstract
The kynurenine pathway (KP) is the dominant pathway for tryptophan degradation in the mammalian body and emerging evidence suggests that acute episodes of sleep deprivation (SD) disrupt tryptophan metabolism via the KP. Increases in the neuroactive KP metabolite kynurenic acid (KYNA) during pregnancy may lead to a higher risk for disrupted neurodevelopment in the offspring. As pregnancy is a critical period during which several factors, including sleep disruptions, could disrupt the fetal environment, we presently explored the relationship between maternal SD and KP metabolism and immune pathways in maternal, placenta, and fetal tissues. Pregnant Wistar rat dams were sleep deprived by gentle handling for 5 h from zeitgeber time (ZT) 0 to ZT 5. Experimental cohorts included: i) controls, ii) one session of SD on embryonic day (ED) 18 or iii) three sessions of SD occurring daily on ED 16, ED 17 and ED 18. Maternal (plasma, brain), placental and fetal (plasma, brain) tissues were collected immediately after the last session of SD or after 24 h of recovery from SD. Respective controls were euthanized at ZT 5 on ED 18 or ED 19. Maternal plasma corticosterone and fetal brain KYNA were significantly elevated only after one session of SD on ED 18. Importantly, maternal plasma corticosterone levels correlated significantly with fetal brain KYNA levels. In addition, placental levels of the proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-6 (IL-6) were increased following maternal SD, suggesting a relationship between placental immune response to SD and fetal brain KYNA accumulation. Collectively, our results demonstrate that sleep loss during the last week of gestation can adversely impact maternal stress, placental immune function, and fetal brain KYNA levels. We introduce KYNA as a novel molecular target influenced by sleep loss during pregnancy. Prenatal sleep deprivation influences kynurenine pathway metabolism in utero. Fetal brain kynurenic acid (KYNA) is elevated after maternal sleep deprivation. Maternal plasma corticosterone is increased after sleep deprivation. Prenatal sleep deprivation induces placental and fetal brain cytokines. These data support an interplay with stress, in utero inflammation, and KYNA.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nickole R Kanyuch
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Casey A Cole
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Homayoun Valafar
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Jessica Deslauriers
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
24
|
Hu Z, Du X, Yang Y, Botchway BOA, Fang M. Progesterone and fluoxetine treatments of postpartum depressive-like behavior in rat model. Cell Biol Int 2019; 43:539-552. [PMID: 30811083 DOI: 10.1002/cbin.11123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/23/2019] [Indexed: 12/19/2022]
Abstract
Research studies have indicated that alterations in plasma progesterone levels might be associated with the hippocampal synaptic plasticity of postpartum depressive-like behavior. Herein, we assess both progesterone and fluoxetine effects in adult female Sprague-Dawley rats with postpartum depressive-like behavior. Depressive-like behavior of postpartum rats was established using chronic ultra-mild stress (CUMS) method for 1 week from gestation day 15. Postpartum rats that showed depressive-like behavior were treated with either progesterone (subcutaneously, 0.5 mg/kg) from gestation day 17 to gestation day 22 or fluoxetine (by gavage, 10 mg/kg/day) for 4 weeks after birth. Open field and sucrose preference tests were conducted at the start, week 2 and week 4 postpartum. Golgi staining, immunofluorescence and Western blot analyses of rats' hippocampi were conducted on week 4 postpartum. Results showed CUMS increases depressive-like behavior, however, treatment with progesterone and fluoxetine improves this behavior. Both progesterone and fluoxetine treatments increase the numbers of dendritic spines pyramidal neurons in the CA3 region of the hippocampus as well as protein expression levels of microtubule-associated protein 2 (MAP-2) and synaptophysin (SYP). CUMS-induced decrement of MAP-2 and SYP protein expressions can be prevented by treatment with progesterone in advanced pregnant stage and fluoxetine in the postpartum period.
Collapse
Affiliation(s)
- Zhiying Hu
- Hangzhou Red Cross Hospital, Hangzhou, China
| | - Xiaoxue Du
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yang Yang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
25
|
Immune Challenge Alters Reactivity of Hippocampal Noradrenergic System in Prenatally Stressed Aged Mice. Neural Plast 2019; 2019:3152129. [PMID: 30804990 PMCID: PMC6360630 DOI: 10.1155/2019/3152129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
Prenatal stress (PS) has long-term sequelae for the morphological and functional status of the central nervous system of the progeny. A PS-induced proinflammatory status of the organism may result in an impairment of both hippocampal synaptic plasticity and hippocampus-dependent memory formation in adults. We addressed here the question of how PS-induced alterations in the immune response in young and old mice may contribute to changes in hippocampal function in aging. Immune stimulation (via LPS injection) significantly affected the ability of the hippocampal CA3-CA1 synapse of PS mice to undergo long-term potentiation (LTP). Elevated corticosterone level in the blood of aged PS mice that is known to influence LTP magnitude indicates a chronic activation of the HPA axis due to the in utero stress exposure. We investigated the contribution of adrenergic receptors to the modulation of hippocampal synaptic plasticity of aged mice and found that impaired LTP in the PS-LPS group was indeed rescued by application of isoproterenol (a nonspecific noradrenergic agonist). Further exploration of the mechanisms of the observed phenomena will add to our understanding of the interaction between PS and proinflammatory immune activation and its contribution to the functional and structural integrity of the aging brain.
Collapse
|
26
|
Maternal stress during pregnancy induces depressive-like behavior only in female offspring and correlates to their hippocampal Avp and Oxt receptor expression. Behav Brain Res 2018; 353:1-10. [DOI: 10.1016/j.bbr.2018.06.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
|
27
|
Schepanski S, Buss C, Hanganu-Opatz IL, Arck PC. Prenatal Immune and Endocrine Modulators of Offspring's Brain Development and Cognitive Functions Later in Life. Front Immunol 2018; 9:2186. [PMID: 30319639 PMCID: PMC6168638 DOI: 10.3389/fimmu.2018.02186] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022] Open
Abstract
Milestones of brain development in mammals are completed before birth, which provide the prerequisite for cognitive and intellectual performances of the offspring. Prenatal challenges, such as maternal stress experience or infections, have been linked to impaired cognitive development, poor intellectual performances as well as neurodevelopmental and psychiatric disorders in the offspring later in life. Fetal microglial cells may be the target of such challenges and could be functionally modified by maternal markers. Maternal markers can cross the placenta and reach the fetus, a phenomenon commonly referred to as “vertical transfer.” These maternal markers include hormones, such as glucocorticoids, and also maternal immune cells and cytokines, all of which can be altered in response to prenatal challenges. Whilst it is difficult to discriminate between the maternal or fetal origin of glucocorticoids and cytokines in the offspring, immune cells of maternal origin—although low in frequency—can be clearly set apart from offspring's cells in the fetal and adult brain. To date, insights into the functional role of these cells are limited, but it is emergingly recognized that these maternal microchimeric cells may affect fetal brain development, as well as post-natal cognitive performances and behavior. Moreover, the inheritance of vertically transferred cells across generations has been proposed, yielding to the presence of a microchiome in individuals. Hence, it will be one of the scientific challenges in the field of neuroimmunology to identify the functional role of maternal microchimeric cells as well as the brain microchiome. Maternal microchimeric cells, along with hormones and cytokines, may induce epigenetic changes in the fetal brain. Recent data underpin that brain development in response to prenatal stress challenges can be altered across several generations, independent of a genetic predisposition, supporting an epigenetic inheritance. We here discuss how fetal brain development and offspring's cognitive functions later in life is modulated in the turnstile of prenatal challenges by introducing novel and recently emerging pathway, involving maternal hormones and immune markers.
Collapse
Affiliation(s)
- Steven Schepanski
- Laboratory of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Buss
- Institute of Medical Psychology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Development, Health, and Disease Research Program, University of California, Irvine, Orange, CA, United States
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C Arck
- Laboratory of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
28
|
Lesuis SL, Hoeijmakers L, Korosi A, de Rooij SR, Swaab DF, Kessels HW, Lucassen PJ, Krugers HJ. Vulnerability and resilience to Alzheimer's disease: early life conditions modulate neuropathology and determine cognitive reserve. Alzheimers Res Ther 2018; 10:95. [PMID: 30227888 PMCID: PMC6145191 DOI: 10.1186/s13195-018-0422-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a high prevalence among the elderly and a huge personal and societal impact. Recent epidemiological studies have indicated that the incidence and age of onset of sporadic AD can be modified by lifestyle factors such as education, exercise, and (early) stress exposure. Early life adversity is known to promote cognitive decline at a later age and to accelerate aging, which are both primary risk factors for AD. In rodent models, exposure to 'negative' or 'positive' early life experiences was recently found to modulate various measures of AD neuropathology, such as amyloid-beta levels and cognition at later ages. Although there is emerging interest in understanding whether experiences during early postnatal life also modulate AD risk in humans, the mechanisms and possible substrates underlying these long-lasting effects remain elusive. METHODS We review literature and discuss the role of early life experiences in determining later age and AD-related processes from a brain and cognitive 'reserve' perspective. We focus on rodent studies and the identification of possible early determinants of later AD vulnerability or resilience in relation to early life adversity/enrichment. RESULTS Potential substrates and mediators of early life experiences that may influence the development of AD pathology and cognitive decline are: programming of the hypothalamic-pituitary-adrenal axis, priming of the neuroinflammatory response, dendritic and synaptic complexity and function, overall brain plasticity, and proteins such as early growth response protein 1 (EGR1), activity regulated cytoskeleton-associated protein (Arc), and repressor element-1 silencing transcription factor (REST). CONCLUSIONS We conclude from these rodent studies that the early postnatal period is an important and sensitive phase that influences the vulnerability to develop AD pathology. Yet translational studies are required to investigate whether early life experiences also modify AD development in human studies, and whether similar molecular mediators can be identified in the sensitivity to develop AD in humans.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Susanne R. de Rooij
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics & Bio informatics, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Dick F. Swaab
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Helmut W. Kessels
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Department of Cellular and Computational Neuroscience, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
29
|
On the Developmental Timing of Stress: Delineating Sex-Specific Effects of Stress across Development on Adult Behavior. Brain Sci 2018; 8:brainsci8070121. [PMID: 29966252 PMCID: PMC6071226 DOI: 10.3390/brainsci8070121] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Stress, and the chronic overactivation of major stress hormones, is associated with several neuropsychiatric disorders. However, clinical literature on the exact role of stress either as a causative, triggering, or modulatory factor to mental illness remains unclear. We suggest that the impact of stress on the brain and behavior is heavily dependent on the developmental timing at which the stress has occurred, and as such, this may contribute to the overall variability reported on the association of stress and mental illness. Here, animal models provide a way to comprehensively assess the temporal impact of stress on behavior in a controlled manner. This review particularly focuses on the long-term impact of stress on behavior in various rodent stress models at three major developmental time points: early life, adolescence, and adulthood. We characterize the various stressor paradigms into physical, social, and pharmacological, and discuss commonalities and differences observed across these various stress-inducing methods. In addition, we discuss here how sex can influence the impact of stress at various developmental time points. We conclude here that early postnatal life and adolescence represent particular periods of vulnerability, but that stress exposure during early life can sometimes lead to resilience, particularly to fear-potentiated memories. In the adult brain, while shorter periods of stress tended to enhance spatial memory, longer periods caused impairments. Overall, males tended to be more vulnerable to the long-term effects of early life and adolescent stress, albeit very few studies incorporate both sexes, and further well-powered sex comparisons are needed.
Collapse
|
30
|
Wolf IAC, Gilles M, Peus V, Scharnholz B, Seibert J, Jennen-Steinmetz C, Krumm B, Rietschel M, Deuschle M, Laucht M. Impact of prenatal stress on mother-infant dyadic behavior during the still-face paradigm. Borderline Personal Disord Emot Dysregul 2018; 5:2. [PMID: 29403645 PMCID: PMC5778796 DOI: 10.1186/s40479-018-0078-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/03/2018] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Mother-infant interaction provides important training for the infant's ability to cope with stress and the development of resilience. Prenatal stress (PS) and its impact on the offspring's development have long been a focus of stress research, with studies highlighting both harmful and beneficial effects. The aim of the current study was to examine the possible influence of both psychological stress and hypothalamic-pituitary-adrenal (HPA) axis activity during pregnancy with mother-child dyadic behavior following stress exposure. METHODS The behavior of 164 mother-infant dyads during the still-face situation was filmed at six months postpartum and coded into three dyadic patterns: 1) both positive, 2) infant protesting-mother positive, and 3) infant protesting-mother negative. PS exposure was assessed prenatally according to psychological measures (i.e., psychopathological, perceived and psychosocial PS; n = 164) and HPA axis activity measures (maternal salivary cortisol, i.e., cortisol decline and area under the curve with respect to ground (AUCg); n = 134). RESULTS Mother-infant dyads in both the high- and low-stress groups showed decreasing positive and increasing negative dyadic behavior in the reunion episode, which is associated with the well-known "still-face" and "carry-over" effect. Furthermore, mother-infant dyads with higher psychosocial PS exhibited significantly more positive dyadic behavior than the low psychosocial PS group in the first play episode, but not in the reunion episode. Similarly, mother-infant dyads with high HPA axis activity (i.e. high AUCg) but steeper diurnal cortisol decline (i.e. cortisol decline) displayed significantly less negative behavior in the reunion episode than dyads with low HPA axis activity. No significant results were found for psychopathological stress and perceived stress. CONCLUSIONS The results suggest a beneficial effect of higher psychosocial PS and higher prenatal maternal HPA axis activity in late gestation, which is in line with "stress inoculation" theories.
Collapse
Affiliation(s)
- Isabell Ann-Cathrin Wolf
- 1Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim / Heidelberg University, J 5, 68159 Mannheim, Germany
| | - Maria Gilles
- 1Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim / Heidelberg University, J 5, 68159 Mannheim, Germany
| | - Verena Peus
- 1Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim / Heidelberg University, J 5, 68159 Mannheim, Germany
| | - Barbara Scharnholz
- 1Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim / Heidelberg University, J 5, 68159 Mannheim, Germany
| | - Julia Seibert
- Clinic for General Psychiatry, Center for Psychosocial Medicine, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Christine Jennen-Steinmetz
- 3Department of Biostatistics, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bertram Krumm
- 3Department of Biostatistics, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcella Rietschel
- 4Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Deuschle
- 1Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim / Heidelberg University, J 5, 68159 Mannheim, Germany
| | - Manfred Laucht
- 5Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,6Department of Psychology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
31
|
Impact of prenatal stress on the dyadic behavior of mothers and their 6-month-old infants during a play situation: role of different dimensions of stress. J Neural Transm (Vienna) 2017; 124:1251-1260. [DOI: 10.1007/s00702-017-1770-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/23/2017] [Indexed: 01/13/2023]
|
32
|
Maschietto M, Bastos LC, Tahira AC, Bastos EP, Euclydes VLV, Brentani A, Fink G, de Baumont A, Felipe-Silva A, Francisco RPV, Gouveia G, Grisi SJFE, Escobar AMU, Moreira-Filho CA, Polanczyk GV, Miguel EC, Brentani H. Sex differences in DNA methylation of the cord blood are related to sex-bias psychiatric diseases. Sci Rep 2017; 7:44547. [PMID: 28303968 PMCID: PMC5355991 DOI: 10.1038/srep44547] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/10/2017] [Indexed: 12/19/2022] Open
Abstract
Sex differences in the prevalence of psychiatric disorders are well documented, with exposure to stress during gestation differentially impacting females and males. We explored sex-specific DNA methylation in the cord blood of 39 females and 32 males born at term and with appropriate weight at birth regarding their potential connection to psychiatric outcomes. Mothers were interviewed to gather information about environmental factors (gestational exposure) that could interfere with the methylation profiles in the newborns. Bisulphite converted DNA was hybridized to Illumina HumanMethylation450 BeadChips. Excluding XYS probes, there were 2,332 differentially methylated CpG sites (DMSs) between sexes, which were enriched within brain modules of co-methylated CpGs during brain development and also differentially methylated in the brains of boys and girls. Genes associated with the DMSs were enriched for neurodevelopmental disorders, particularly for CpG sites found differentially methylated in brain tissue between patients with schizophrenia and controls. Moreover, the DMS had an overlap of 890 (38%) CpG sites with a cohort submitted to toxic exposition during gestation. This study supports the evidences that sex differences in DNA methylation of autosomes act as a primary driver of sex differences that are found in psychiatric outcomes.
Collapse
Affiliation(s)
- Mariana Maschietto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | | | | | | | | | - Alexandra Brentani
- Department of Pediatrics, University of São Paulo Medical School, SP, Brazil
| | - Günther Fink
- Department of Global Health and Population, Harvard School of Public Health, USA
| | | | | | | | - Gisele Gouveia
- Institute of Psychiatry, University of São Paulo Medical School, SP, Brazil
| | | | | | | | | | | | - Helena Brentani
- Institute of Psychiatry, University of São Paulo Medical School, SP, Brazil
| |
Collapse
|
33
|
Suzuki A, Iinuma M, Hayashi S, Sato Y, Azuma K, Kubo KY. Maternal chewing during prenatal stress ameliorates stress-induced hypomyelination, synaptic alterations, and learning impairment in mouse offspring. Brain Res 2016; 1651:36-43. [PMID: 27613358 DOI: 10.1016/j.brainres.2016.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/12/2016] [Accepted: 09/05/2016] [Indexed: 01/24/2023]
Abstract
Maternal chewing during prenatal stress attenuates both the development of stress-induced learning deficits and decreased cell proliferation in mouse hippocampal dentate gyrus. Hippocampal myelination affects spatial memory and the synaptic structure is a key mediator of neuronal communication. We investigated whether maternal chewing during prenatal stress ameliorates stress-induced alterations of hippocampal myelin and synapses, and impaired development of spatial memory in adult offspring. Pregnant mice were divided into control, stress, and stress/chewing groups. Stress was induced by placing mice in a ventilated restraint tube, and was initiated on day 12 of pregnancy and continued until delivery. Mice in the stress/chewing group were given a wooden stick to chew during restraint. In 1-month-old pups, spatial memory was assessed in the Morris water maze, and hippocampal oligodendrocytes and synapses in CA1 were assayed by immunohistochemistry and electron microscopy. Prenatal stress led to impaired learning ability, and decreased immunoreactivity of myelin basic protein (MBP) and 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) in the hippocampal CA1 in adult offspring. Numerous myelin sheath abnormalities were observed. The G-ratio [axonal diameter to axonal fiber diameter (axon plus myelin sheath)] was increased and postsynaptic density length was decreased in the hippocampal CA1 region. Maternal chewing during stress attenuated the prenatal stress-induced impairment of spatial memory, and the decreased MBP and CNPase immunoreactivity, increased G-ratios, and decreased postsynaptic-density length in the hippocampal CA1 region. These findings suggest that chewing during prenatal stress in dams could be an effective coping strategy to prevent hippocampal behavioral and morphologic impairments in their offspring.
Collapse
Affiliation(s)
- Ayumi Suzuki
- Department of Pediatric Dentistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Mitsuo Iinuma
- Department of Pediatric Dentistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Sakurako Hayashi
- Department of Pediatric Dentistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Yuichi Sato
- Department of Molecular Diagnostics, Kitasato University School of Allied Health Science, Kitasato 1-15-1, Minamiku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kagaku Azuma
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555, Japan
| | - Kin-Ya Kubo
- Seijoh University Graduate School of Health Care Studies, 2-172, Fukinodai, Tokai, Aichi 476-8588, Japan.
| |
Collapse
|
34
|
Barbie-Shoshani Y, Shoham S, Bejar C, Weinstock M. Sex-Specific Effects of Prenatal Stress on Memory and Markers of Neuronal Activity in Juvenile Rats. Dev Neurosci 2016; 38:206-219. [DOI: 10.1159/000446981] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 05/20/2016] [Indexed: 11/19/2022] Open
Abstract
Stress during pregnancy can increase the incidence of emotional problems, learning and language difficulties in human infants and pre-adolescents. Most preclinical studies in rats that attempted to find experimental support for these observations were performed in adult male offspring, but the results are inconsistent. The aim of the current study was to examine the effect of prenatal stress on novel object recognition (NOR) and spatial learning and memory in the Morris water maze (MWM) of juvenile rats of both sexes. By the use of fluorescence immunohistochemistry and protein measurements by Western blot, we measured the expression of markers of neurogenesis (doublecortin, DCX) and neuronal activity that are important for synaptic plasticity and learning (c-fos, GluR1, nNOS). Since neuronal activity in the developing and adult brain can be regulated by astrocytes, we also measured the number of astrocytes and the expression of two astroglial proteins (GFAP and S100B) in the stress-responsive hippocampal dentate gyrus (DG). Experiments were performed on littermates of rats in which its effects on behavior were measured. We found for the first time that juvenile females performed better than males in the NOR and MWM tests. They also had higher densities of DCX and c-fos in the DG, together with the expression of nNOS and GluR1 in the subgranular zone (SGZ) of the DG. There were no sex differences in the expression of GFAP and S100B in astrocytes. Prenatal stress did not affect NOR in females, but improved it in males, together with an increase in DCX+ and c-fos, the number of GFAP-expressing astrocytes and the intensity of GFAP and S100B immunofluorescence in the DG. Staining intensity of GluR1 and nNOS in the hilus and SGZ of the DG, and protein expression in the whole DG, was unchanged in prenatally stressed males. Thus, prenatal stress changed the behavior and expression of key proteins in the DG to resemble that in females. A reduction in plasma testosterone, which although not attaining statistical significance was associated with that in anogenital distance, may contribute to the effect of prenatal stress in males. In females, prenatal stress had no effect on c-fos, DCX or the number of astrocytes but reduced the staining intensity of GluR1 and nNOS. Protein expression of nNOS was also significantly lower than that in prenatally stressed males. The differential effects of prenatal stress on hippocampal neuronal and glial markers may help to explain the sex-dependent effect on spatial learning in prepubertal rats.
Collapse
|
35
|
Edwards AM, Cameron EZ, Pereira JC, Wapstra E, Ferguson-Smith MA, Horton SR, Thomasson K. Gestational experience alters sex allocation in the subsequent generation. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160210. [PMID: 27493776 PMCID: PMC4968468 DOI: 10.1098/rsos.160210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/08/2016] [Indexed: 06/06/2023]
Abstract
Empirical tests of adaptive maternal sex allocation hypotheses have presented inconsistent results in mammals. The possibility that mothers are constrained in their ability to adjust sex ratios could explain some of the remaining variation. Maternal effects, the influence of the maternal phenotype or genotype on her developing offspring, may constrain sex allocation through physiological changes in response to the gestational environment. We tested if maternal effects constrain future parental sex allocation through a lowered gestational stress environment in laboratory mice. Females that experienced lowered stress as embryos in utero gave birth to female-biased litters as adults, with no change to litter size. Changes in offspring sex ratio was linked to peri-conceptual glucose, as those females that had increasing blood glucose peri-conceptionally gave birth to litters with a higher male to female sex ratio. There was, however, no effect of the lowered prenatal stress for developing male embryos and their sperm sex ratio when adult. We discuss the implications of maternal effects and maternal stress environment on the lifelong physiology of the offspring, particularly as a constraint on later maternal sex allocation.
Collapse
Affiliation(s)
- A. M. Edwards
- School of Biological Sciences, University of Tasmania, Hobart, Tasmania, Australia
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - E. Z. Cameron
- School of Biological Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | - J. C. Pereira
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Cytocell Ltd., Cambridge Technopark, Newmarket Road, Cambridge, UK
| | - E. Wapstra
- School of Biological Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | | | - S. R. Horton
- School of Biological Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | - K. Thomasson
- School of Biological Sciences, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
36
|
Altered brain morphology and functional connectivity reflect a vulnerable affective state after cumulative multigenerational stress in rats. Neuroscience 2016; 330:79-89. [PMID: 27241944 DOI: 10.1016/j.neuroscience.2016.05.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 01/09/2023]
Abstract
Prenatal stress is a risk factor for abnormal neuroanatomical, cognitive, behavioral and mental health outcomes with potentially transgenerational consequences. Females in general seem more resilient to the effects of prenatal stress than males. Here, we examined if repeated stress across generations may diminish stress resiliency and cumulatively enhance the susceptibility for adverse health outcomes in females. Pregnant female rats of three successive generations were exposed to stress from gestational days 12-18 to generate multigenerational prenatal stress (MPS) in the maternal lineage. Stress response was measured by plasma corticosterone levels and open-field exploration in each generation. Neuromorphological consequences of MPS were investigated in the F3 generation using in vivo manganese-enhanced magnetic resonance imaging (MEMRI), T2-relaxometry, and cytoarchitectonics in relation to candidate gene expression involved in brain plasticity and mental health. Each additional generation of prenatal stress incrementally elevated hypothalamic-pituitary-adrenal axis activation, anxiety-like and aversive behaviors in adult female offspring. Elevated stress responses in the MPS F3 generation were accompanied by reduced neural density in prefrontal cortex, hippocampus and whole brain along with altered brain activation patterns in in vivo MEMRI. MPS increased ephrin receptor A5 (Epha5), neuronal growth regulator (Negr1) and synaptosomal-associated protein 25 (Snap25) gene expression and reduced fibroblast growth factor 12 (Fgf12) in prefrontal cortex. These genes regulate neuronal maturation, arborization and synaptic plasticity and may explain altered brain cytoarchitectonics and connectivity. These findings emphasize that recurrent stress across generations may cumulatively increase stress vulnerability and the risk of adverse health outcomes through perinatal programing in females.
Collapse
|
37
|
Gröger N, Matas E, Gos T, Lesse A, Poeggel G, Braun K, Bock J. The transgenerational transmission of childhood adversity: behavioral, cellular, and epigenetic correlates. J Neural Transm (Vienna) 2016; 123:1037-52. [DOI: 10.1007/s00702-016-1570-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/02/2016] [Indexed: 12/21/2022]
|
38
|
Are there physiological constraints on maternal ability to adjust sex ratios in mammals? J Zool (1987) 2016. [DOI: 10.1111/jzo.12327] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
39
|
Lasting Differential Effects on Plasticity Induced by Prenatal Stress in Dorsal and Ventral Hippocampus. Neural Plast 2016; 2016:2540462. [PMID: 26881096 PMCID: PMC4736977 DOI: 10.1155/2016/2540462] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/18/2015] [Accepted: 12/20/2015] [Indexed: 12/14/2022] Open
Abstract
Early life adversaries have a profound impact on the developing brain structure and functions that persist long after the original traumatic experience has vanished. One of the extensively studied brain structures in relation to early life stress has been the hippocampus because of its unique association with cognitive processes of the brain. While the entire hippocampus shares the same intrinsic organization, it assumes different functions in its dorsal and ventral sectors (DH and VH, resp.), based on different connectivity with other brain structures. In the present review, we summarize the differences between DH and VH and discuss functional and structural effects of prenatal stress in the two sectors, with the realization that much is yet to be explored in understanding the opposite reactivity of the DH and VH to stressful stimulation.
Collapse
|
40
|
Bock J, Wainstock T, Braun K, Segal M. Stress In Utero: Prenatal Programming of Brain Plasticity and Cognition. Biol Psychiatry 2015; 78:315-26. [PMID: 25863359 DOI: 10.1016/j.biopsych.2015.02.036] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/10/2015] [Accepted: 02/25/2015] [Indexed: 12/17/2022]
Abstract
Animal studies confirm earlier anecdotal observations in humans to indicate that early life experience has a profound impact on adult behavior, years after the original experience has vanished. These studies also highlight the role of early life adversaries in the shaping of a disordered brain. Evidence is accumulating to indicate that the epigenome, through which the environment regulates gene expression, is responsible for long-lasting effects of stress during pregnancy on brain and behavior. A possible differential effect of the environment on the epigenome may underlie the observation that only a small fraction of a population with similar genetic background deteriorates into mental disorders. Considerable progress has been made in the untangling of the epigenetic mechanisms that regulate emotional brain development. The present review focuses on the lasting effects of prenatal stress on brain plasticity and cognitive functions in human and rodent models. Although human studies stress the significance of early life experience in functional maturation, they lack the rigor inherent in controlled animal experiments. Furthermore, the analysis of molecular and cellular mechanisms affected by prenatal stress is possible only in experimental animals. The present review attempts to link human and animal studies while proposing molecular mechanisms that interfere with functional brain development.
Collapse
Affiliation(s)
- Joerg Bock
- Otto von Guericke University Magdeburg (JB, KB), Magdeburg, Germany
| | - Tamar Wainstock
- Rollins School of Public Health (TW), Emory University, Atlanta, Georgia
| | - Katharina Braun
- Otto von Guericke University Magdeburg (JB, KB), Magdeburg, Germany
| | - Menahem Segal
- Department of Neurobiology (MS) Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
41
|
Soztutar E, Colak E, Ulupinar E. Gender- and anxiety level-dependent effects of perinatal stress exposure on medial prefrontal cortex. Exp Neurol 2015; 275 Pt 2:274-84. [PMID: 26057948 DOI: 10.1016/j.expneurol.2015.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 01/15/2023]
Abstract
Early life stress leads to psychopathological processes correlated with the predisposition of individuals. Prolonged development of the prefrontal cortex (PFC), playing a critical role in the cognition, personality and social behavior, makes it susceptible to adverse conditions. In this study, we evaluated the dendritic morphology of medial PFC neurons in rats subjected to perinatal stress exposure. Unbiased stereological counting methods showed that total number estimation of c-Fos (+) nuclei, indicating the neuronal activation upon stressful challenge, significantly increased in high anxious animals compared with low anxious and control groups, in both gender. Golgi-Cox staining of neurons displayed anxiety level- and sex-dependent reduction in the dendritic complexity and spine density of pyramidal neurons, especially in the stressed males. While the total length of dendrites were not correlational; density of spines, specifically the mushroom subtypes, showed a negative correlation with the anxiety level of stressed animals. These results suggest that medial PFC is a critical site of neural plasticity within the stressor controllability paradigm. Outcomes of early life stress might be predicted by analyzing the density and morphology of spines in the apical dendrites of pyramidal neurons in correlation with the anxiety-like behavior of animals.
Collapse
Affiliation(s)
- Erdem Soztutar
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey; Interdisciplinary Neuroscience Department, Health Science Institute of Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Ertugrul Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Emel Ulupinar
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey; Interdisciplinary Neuroscience Department, Health Science Institute of Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| |
Collapse
|
42
|
Brunton PJ. Programming the brain and behaviour by early-life stress: a focus on neuroactive steroids. J Neuroendocrinol 2015; 27:468-80. [PMID: 25688636 DOI: 10.1111/jne.12265] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
Animal studies have amply demonstrated that stress exposure during pregnancy or in early postnatal life can adversely influence brain development and have long-term 'programming' effects on future brain function and behaviour. Furthermore, a growing body of evidence from human studies supports the hypothesis that some psychiatric disorders may have developmental origins. Here, the focus is on three adverse consequences of early-life stress: dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, heightened anxiety behaviour and cognitive impairments, with review of what is known about the underlying central mechanisms. Neuroactive steroids modulate neuronal activity and play a key role in neurodevelopment. Moreover they can negatively modulate activity of the HPA axis, exert anxiolytic actions and influence cognitive performance. Thus, neuroactive steroids may provide a link between early-life stress and the resultant adverse effects on the brain and behaviour. Here, a role for neuroactive steroids, in particular the 5α-reduced/3α-hydroxylated metabolites of progesterone, testosterone and deoxycorticosterone, is discussed in the context of early-life stress. Furthermore, the impact of early-life stress on the brain's capacity to generate neurosteroids is considered and the evidence for an ability of neuroactive steroids to over-write the negative effects of early-life stress on the brain and behaviour is examined. An enhanced understanding of the influence of early-life stress on brain neurosteroid systems could aid the identification of new targets for developing treatments for stress-related conditions in humans.
Collapse
Affiliation(s)
- P J Brunton
- Division of Neurobiology, The Roslin Institute & R(D)SVS, University of Edinburgh, Midlothian, UK
| |
Collapse
|
43
|
Changes induced by prenatal stress in behavior and brain morphology: can they be prevented or reversed? ADVANCES IN NEUROBIOLOGY 2015; 10:3-25. [PMID: 25287533 DOI: 10.1007/978-1-4939-1372-5_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This chapter presents a critical analysis of the behavioral alterations reported in the offspring of women exposed to stress and/or depression during pregnancy and the neurochemical and structural changes underlying them. Among the alterations attributed to prenatal stress in humans and experimental rats of both sexes is impaired regulation of the hypothalamic-pituitary-adrenal (HPA) axis, anxiety and exaggerated fear of novelty, and decreased social interaction. Learning and attention deficits are more prevalent in boys and male rats. Fear of novelty and anxiety are associated with enlargement of the amygdala and its corticotropin-releasing factor content, and decreased socialization, with lower oxytocin activity in the amygdala. Learning deficits are associated with a decrease in neurogenesis, dendritic complexity, and spine number in the dorsal hippocampus. Fostering prenatally stressed (PS) pups onto control mothers prevents the dysregulation of the HPA axis and heightened anxiety, indicating a role for postnatal factors in their etiology. By contrast, learning impairment and decreased socialization are not affected by this fostering procedure and are therefore prenatally mediated.In spite of their widespread use in depressed pregnant women, selective serotonin reuptake inhibitor (SSRI) antidepressants do not normalize the behavior of their children. When administered during gestation to stressed rats, SSRIs do not reduce anxiety or learning deficits in their offspring. Moreover, when given to unstressed mothers, SSRIs induce anxiety in the offspring. The detrimental effect of SSRIs may result from inhibition of the serotonin transporter exposing the brain to excess amounts of 5-hydroxytryptamine (5-HT) at a critical time during fetal development.
Collapse
|
44
|
Transgenerational sex-specific impact of preconception stress on the development of dendritic spines and dendritic length in the medial prefrontal cortex. Brain Struct Funct 2014; 221:855-63. [PMID: 25395153 DOI: 10.1007/s00429-014-0940-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/01/2014] [Indexed: 10/24/2022]
Abstract
Perinatal adverse experience programs social and emotional behavioral traits and is a major risk factor for the development of behavioral and psychiatric disorders. Little information is available on how adversity to the mother prior to her first pregnancy (preconception stress, PCS) may affect brain structural development, which may underlie behavioral dysfunction in the offspring. Moreover, little is known about possible sex-dependent consequences of PCS in the offspring. This study examined spine number/density and dendritic length/complexity of layer II/III pyramidal neurons in the anterior cingulate (ACd), prelimbic/infralimbic (PL/IL) and orbitofrontal cortex (OFC) of male and female rats born to mothers exposed to unpredictable variable stress at different time points prior to reproduction. Our main findings are that in line with our hypothesis adversity to the mother before her pregnancy results in highly complex changes in neuronal morphology in the medial prefrontal, but not in the orbitofrontal cortical regions of her future offspring that persist into adulthood. Moreover, our study revealed that (1) in the PCS2 group (offspring of dams mated two weeks after stress) spine numbers and dendritic length and complexity were increased in response to PCS in the ACd and PL/IL, (2) these regional effects depended on the temporal proximity of adversity and conception, (3) in the ACd of the PCS2 group only males and the left hemispheres were affected. We speculate that these transgenerational brain structural changes are mediated by stress-induced epigenetic (re)programming of future gene activity in the oocyte.
Collapse
|
45
|
Chohan TW, Boucher AA, Spencer JR, Kassem MS, Hamdi AA, Karl T, Fok SY, Bennett MR, Arnold JC. Partial genetic deletion of neuregulin 1 modulates the effects of stress on sensorimotor gating, dendritic morphology, and HPA axis activity in adolescent mice. Schizophr Bull 2014; 40:1272-84. [PMID: 24442851 PMCID: PMC4193694 DOI: 10.1093/schbul/sbt193] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stress has been linked to the pathogenesis of schizophrenia. Genetic variation in neuregulin 1 (NRG1) increases the risk of developing schizophrenia and may help predict which high-risk individuals will transition to psychosis. NRG1 also modulates sensorimotor gating, a schizophrenia endophenotype. We used an animal model to demonstrate that partial genetic deletion of Nrg1 interacts with stress to promote neurobehavioral deficits of relevance to schizophrenia. Nrg1 heterozygous (HET) mice displayed greater acute stress-induced anxiety-related behavior than wild-type (WT) mice. Repeated stress in adolescence disrupted the normal development of higher prepulse inhibition of startle selectively in Nrg1 HET mice but not in WT mice. Further, repeated stress increased dendritic spine density in pyramidal neurons of the medial prefrontal cortex (mPFC) selectively in Nrg1 HET mice. Partial genetic deletion of Nrg1 also modulated the adaptive response of the hypothalamic-pituitary-adrenal axis to repeated stress, with Nrg1 HET displaying a reduced repeated stress-induced level of plasma corticosterone than WT mice. Our results demonstrate that Nrg1 confers vulnerability to repeated stress-induced sensorimotor gating deficits, dendritic spine growth in the mPFC, and an abberant endocrine response in adolescence.
Collapse
Affiliation(s)
- Tariq W. Chohan
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia;,Discipline of Pharmacology, School of Medical Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Aurelie A. Boucher
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Jarrah R. Spencer
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia;,Discipline of Pharmacology, School of Medical Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Mustafa S. Kassem
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Areeg A. Hamdi
- Discipline of Pharmacology, School of Medical Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Tim Karl
- Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Sandra Y. Fok
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Maxwell R. Bennett
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathon C. Arnold
- The Brain and Mind Research Institute, University of Sydney, Sydney, NSW 2006, Australia;,Discipline of Pharmacology, School of Medical Science, University of Sydney, Sydney, NSW 2006, Australia;,*To whom correspondence should be addressed; The Brain and Mind Research Institute, University of Sydney, 94-100 Mallett Street, Sydney, Australia; tel: +61-2-9351-0812, e-mail:
| |
Collapse
|
46
|
Ulupinar E, Erol K, Ay H, Yucel F. Rearing conditions differently affect the motor performance and cerebellar morphology of prenatally stressed juvenile rats. Behav Brain Res 2014; 278:235-43. [PMID: 25315128 DOI: 10.1016/j.bbr.2014.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/26/2014] [Accepted: 10/02/2014] [Indexed: 02/05/2023]
Abstract
The cerebellum is one of the most vulnerable parts of the brain to environmental changes. In this study, the effect of diverse environmental rearing conditions on the motor performances of prenatally stressed juvenile rats and its reflection to the cerebellar morphology were investigated. Prenatally stressed Wistar rats were grouped according to different rearing conditions (Enriched=EC, Standard=SC and Isolated=IC) after weaning. Six weeks later, male and female offspring from different litters were tested behaviorally. In rotarod and string suspension tests, females gained better scores than males. Significant gender and housing effects were observed especially on the motor functions requiring fine skills with the best performance by enriched females, but the worst by enriched males. The susceptibility of cerebellar macro- and micro-neurons to environmental conditions was compared using stereological methods. In female groups, no differences were observed in the volume proportions of cerebellar layers, soma sizes and the numerical densities of granule or Purkinje cells. However, a significant interaction between housing and gender was observed in the granule to Purkinje cell ratio of males, due to the increased numerical densities of the granule cells in enriched males. These data imply that proper functioning of the cerebellum relies on its well organized and evolutionarily conserved structure and circuitry. Although early life stress leads to long term behavioral and neurobiological consequences in the offspring, diverse rearing conditions can alter the motor skills of animals and synaptic connectivity between Purkinje and granular cells in a gender dependent manner.
Collapse
Affiliation(s)
- Emel Ulupinar
- Department of Anatomy, Faculty of Medicine, Eskişehir Osmangazi University, 26040 Eskisehir, Turkey; Interdisciplinary Neuroscience Department, Health Science Institute of Eskişehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Kevser Erol
- Interdisciplinary Neuroscience Department, Health Science Institute of Eskişehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Pharmacology, Faculty of Medicine, Eskişehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hakan Ay
- Department of Anatomy, Faculty of Medicine, Eskişehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Ferruh Yucel
- Department of Anatomy, Faculty of Medicine, Eskişehir Osmangazi University, 26040 Eskisehir, Turkey; Interdisciplinary Neuroscience Department, Health Science Institute of Eskişehir Osmangazi University, 26040 Eskisehir, Turkey
| |
Collapse
|
47
|
Barzegar M, Sajjadi FS, Talaei SA, Hamidi G, Salami M. Prenatal exposure to noise stress: Anxiety, impaired spatial memory, and deteriorated hippocampal plasticity in postnatal life. Hippocampus 2014; 25:187-96. [DOI: 10.1002/hipo.22363] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/23/2014] [Accepted: 09/05/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Marzieh Barzegar
- Physiology Research Center; Kashan University of Medical Sciences; Kashan Islamic Republic of Iran
| | - Fatemeh Sadat Sajjadi
- Physiology Research Center; Kashan University of Medical Sciences; Kashan Islamic Republic of Iran
| | - Sayyed Alireza Talaei
- Physiology Research Center; Kashan University of Medical Sciences; Kashan Islamic Republic of Iran
| | - Gholamali Hamidi
- Physiology Research Center; Kashan University of Medical Sciences; Kashan Islamic Republic of Iran
| | - Mahmoud Salami
- Physiology Research Center; Kashan University of Medical Sciences; Kashan Islamic Republic of Iran
| |
Collapse
|
48
|
Gröger N, Bock J, Goehler D, Blume N, Lisson N, Poeggel G, Braun K. Stress in utero alters neonatal stress-induced regulation of the synaptic plasticity proteins Arc and Egr1 in a sex-specific manner. Brain Struct Funct 2014; 221:679-85. [PMID: 25239865 DOI: 10.1007/s00429-014-0889-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/09/2014] [Indexed: 12/23/2022]
Abstract
The present study in juvenile rats investigated a "two-hit model" to test the impact of prenatal stress exposure ("first hit") on the regulation of the synaptic plasticity immediate early genes Arc and Egr1 in response to a second neonatal stressor ("second hit") in a sex-specific manner. Three stress-exposed animal groups were compared at the age of 21 days in relation to unstressed controls (CON): preS animals were exposed to various unpredictable stressors during the last gestational trimester; postS animals were exposed to 45 min restraint stress at postnatal day 21, pre/postS animals were exposed to a combination of pre- and postnatal stress as described for the two previous groups. The postS and pre/postS groups were killed 2 h after exposure to the postnatal stressor, males and females were separately analyzed. In line with our hypothesis we detected sex-specific stress sensitivity for both analyzed proteins. Males did not show any significant changes in Arc expression irrespective of the stress condition. In contrast, females, which had been pre-exposed to prenatal stress, displayed an "amplified" Arc upregulation in response to postnatal stress (pre/postS group) compared to unstressed controls, which may reflect a "sensitization" effect of prenatal stress. For Egr1, the females did not show any stress-induced regulation irrespective of the stress condition, whereas in males, which were pre-exposed to prenatal stress, we observed a "protective" effect of prenatal stress on postnatal stress-induced downregulation of Egr1 expression (pre/postS group), which may indicate that prenatal stress exposure may induce "resilience".
Collapse
Affiliation(s)
- Nicole Gröger
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany.
| | - Joerg Bock
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Daniela Goehler
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Nicole Blume
- Institute for Biology, Human Biology, University of Leipzig, Talstraße 33, 04103, Leipzig, Germany
| | - Nicole Lisson
- Institute for Biology, Human Biology, University of Leipzig, Talstraße 33, 04103, Leipzig, Germany
| | - Gerd Poeggel
- Institute for Biology, Human Biology, University of Leipzig, Talstraße 33, 04103, Leipzig, Germany
| | - Katharina Braun
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
49
|
Expression of neurogranin in hippocampus of rat offspring exposed to restraint stress and pulsed magnetic fields. Brain Res 2014; 1570:26-34. [DOI: 10.1016/j.brainres.2014.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/28/2014] [Accepted: 05/02/2014] [Indexed: 11/23/2022]
|
50
|
Fujimoto T, Kubo K, Nishikawa Y, Aou S. Brief neonatal handling alters sexually dimorphic behaviors in adult rats. J Integr Neurosci 2014; 13:61-70. [PMID: 24738539 DOI: 10.1142/s0219635214500046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Several effects of neonatal handling on brain and behavior have been reported. We investigated the effects of neonatal handling on behaviors that have been shown to be sexually dimorphic in rats using an open-field test. "Gender differences" were observed in locomotor activity, exploratory behavior and grooming in the handled group. However, clear gender differences in these behaviors were not observed in the non-handled group. Our findings show that brief daily handling sessions (~ 1 min) in the first 2 weeks of postnatal life increased locomotor activity and exploratory behavior, and that these effects were more pronounced in females. Moreover, many rats in the non-handling group exhibited an increase in defecation relative to the handling group during the 10-min observation period. This suggests that the non-handling group experienced more stress in response to the novel open-field arena, and that this resulted in the absence of gender differences. Notably, this anxiety-related response was attenuated by neonatal handling. Our study underscores the impact of brief neonatal handling on sexually dimorphic behaviors, and indicates that caution should be exercised in controlling for the effects of handling between experimental groups, particularly in neurotoxicological studies that evaluate gender differences.
Collapse
Affiliation(s)
- Tetsuya Fujimoto
- Department of Physiology, Osaka Dental University, Hirakata 573-1121, Japan
| | | | | | | |
Collapse
|