1
|
Peralta-Vallejo N, Cañete T, Sampedro-Viana D, Güell-Falgueras P, Río-Álamos C, Oliveras I, Tobeña A, Aznar S, Fernández-Teruel A. Neonatal handling enhances behavioural and attentional domains, and frontocortical synaptic maturation in rat models of schizophrenia-like behaviour and anxiety-related responses. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111364. [PMID: 40233871 DOI: 10.1016/j.pnpbp.2025.111364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/06/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
The Roman inbred rat strains are a neurodevelopmental model, with the Roman High Avoidance (RHA) presenting specific behaviours and frontal cortex (FC) gene expression changes relevant to schizophrenia symptoms. We wanted to assess the potentially positive modulatory and enduring effects of neonatal handling (NH) on the innate traits associated with both the RHA and their counterpart Roman Low Avoidance (RLA). Male rats received NH or were left untreated (controls). Two different age groups were considered: adolescent and adults. The assessment encompassed exploratory behaviour, social behaviour, anxiety-related behaviour (self-grooming), sensorimotor gating (prepulse inhibition; PPI), and the analysis of gene expression associated with synaptic processes, cortical maturation, and neuroplasticity in the FC. In adolescent rats, NH increased novelty exploration and activity, and reduced novelty-induced self-grooming in RLAs, whereas it improved PPI in RHAs. In adult rats, NH increased novelty-induced activity in both strains, reduced self-grooming in RLA rats, and enhanced social interaction and PPI in RHAs. NH produced significant effects on gene expression in adolescent RHA rats. These effects were observed at the presynaptic level by a reduction of Snap25 and increases of Cables1 and Cdk5, and at the postsynaptic level by increases of Grin2b, Homer1 and Nrg1, as well as by a NH-induced enhancement of Bdnf. NH also increased Nrg1 and Bdnf expression in adult RLA rats. These findings show for the first time that NH is able to modulate several genetically linked synaptic/neuroplasticity alterations in RHA vs. RLA rats, which are paralleled by NH-induced improvements in novelty exploration, social behaviour and sensorimotor gating (PPI).
Collapse
Affiliation(s)
- Natalia Peralta-Vallejo
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain; Centre for Neuroscience and Stereology, and Center for Translational Research, Copenhagen University Hospital Bispebjerg-Frederiksberg, Denmark
| | - Toni Cañete
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Daniel Sampedro-Viana
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Pau Güell-Falgueras
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Cristóbal Río-Álamos
- Department of Psychology, School of Medicine, Austral University of Chile, Valdivia, Chile
| | - Ignasi Oliveras
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain; Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Adolf Tobeña
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Susana Aznar
- Centre for Neuroscience and Stereology, and Center for Translational Research, Copenhagen University Hospital Bispebjerg-Frederiksberg, Denmark.
| | - Alberto Fernández-Teruel
- Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Faculty of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
2
|
García-Saucedo B, Romero-Morales L, Álvarez-Rodríguez C, Cárdenas-Vázquez R, Ávila-Costa MR, Luis J. Father's Absence in the Mongolian gerbil (Meriones unguiculatus) is associated with alterations in paternal behavior, T, cort, presence of ERα, and AR in mPOA/ BNST. Behav Brain Res 2024; 465:114928. [PMID: 38432301 DOI: 10.1016/j.bbr.2024.114928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/12/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Testosterone (T), estrogen receptor alpha (ERα), and androgen receptor (AR) play a significant role in the regulation of paternal behavior. We determined the effects of deprivation of paternal care on alterations in paternal behavior, T concentrations in plasma, and the presence of ERα and AR in the medial preoptic area (mPOA), bed nucleus of the stria terminalis (BNST), medial amygdala (MeA), and olfactory bulb (OB), as well as the corticosterone (CORT) concentrations in plasma caused by deprivation of paternal care in the Mongolian gerbil (Meriones unguiculatus). Twenty pairs of gerbils were formed; the pups were deprived of paternal care (DPC) in 10 pairs. In another 10 pairs, the pups received paternal care (PC). Ten males raised in DPC condition and 10 males raised in PC conditions were mated with virgin females. When they became fathers, each DPC male and PC male was subjected to tests of paternal behavior on day three postpartum. Blood samples were obtained to quantify T and CORT concentrations, and the brains were removed for ERα and AR immunohistochemistry analyses. DPC males gave less care to their pups than PC males, and they had significantly lower T concentrations and levels of ERα and AR in the mPOA and BNST than PC males. DPC males also had higher CORT concentrations than PC males. These results suggest that in the Mongolian gerbil father's absence causes a decrease in paternal care in the offspring, which is associated with alterations in the neuroendocrine mechanisms that regulate it.
Collapse
Affiliation(s)
- Brenda García-Saucedo
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, 7 Circuitos de Posgrado, Ciudad Universitaria, Coyoacán, C.P. 04510, Ciudad de 8 México, México
| | - Luis Romero-Morales
- Laboratorio de Biología de la Reproducción, UMF, FES Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla, Edo. de México, Mexico
| | - Carmen Álvarez-Rodríguez
- Laboratorio de Biología de la Reproducción, UMF, FES Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla, Edo. de México, Mexico
| | - René Cárdenas-Vázquez
- Laboratorio de Biología Animal Experimental, Depto. de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - María Rosa Ávila-Costa
- Laboratorio de Neuromorfología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. Mex, Mexico
| | - Juana Luis
- Laboratorio de Biología de la Reproducción, UMF, FES Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla, Edo. de México, Mexico.
| |
Collapse
|
3
|
da Cunha Nones DC, Novais CO, Rojas VCT, de Paula Franco P, da Silva Estevam E, Silva MS, Giusti-Paiva A, Dos Anjos-Garcia T, Vilela FC. Litter reduction-induced obesity promotes early depressive-like behavior and elevated prefrontal cortex GFAP expression in male offspring. Behav Brain Res 2024; 461:114839. [PMID: 38154508 DOI: 10.1016/j.bbr.2023.114839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
AIMS The present study was developed to investigate how litter reduction-induced obesity promotes early depressive-related behaviors in rodent offspring. MAIN METHODS We employed a standardized litter size reduction protocol, dividing litters into groups: normal litters (NL), consisting of six males and six females pups and small litters (SL), comprising two males and two females pups. Maternal behavior was monitored during the initial week of lactation. Subsequently, we assessed the pups for weight gain, locomotor activity, social play behavior, and performance in forced swimming test. We further evaluated the weights of retroperitoneal and perigonadal fat tissues, along with the expression of glial fibrillary acidic pprotein (GFAP) in the hippocampus and prefrontal cortex of the offspring. KEY FINDINGS Our results indicated that litter size reduction led to an increased the maternal behavior. In contrast, offspring from the SL group displayed greater weight gain and increased, retroperitoneal and perigonadal fat. Both male and female rodents in the SL group exhibited decreased social play behavior, and male offspring spent more time immobile during the forced swimming test, suggesting a depressive-like phenotype. Notably, we observed an increase in the GFAP expression in the prefrontal cortex of male rodents, with a trend toward increased expression in the hippocampus. SIGNIFICANCE Obesity may facilitate the development of early depressive-like behaviors, potentially associated with elevated GFAP expression in the prefrontal cortex.
Collapse
Affiliation(s)
- Débora Cristina da Cunha Nones
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Cíntia Onofra Novais
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Viviana Carolina Trujillo Rojas
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Priscila de Paula Franco
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Elisa da Silva Estevam
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Mariana Santos Silva
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Alexandre Giusti-Paiva
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas da Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Tayllon Dos Anjos-Garcia
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil.
| | - Fabiana Cardoso Vilela
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Centro de Inovação e Ensaios Pré-Clínicos (CIEnP), Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
4
|
Alberry B, Silveira PP. Brain insulin signaling as a potential mediator of early life adversity effects on physical and mental health. Neurosci Biobehav Rev 2023; 153:105350. [PMID: 37544390 DOI: 10.1016/j.neubiorev.2023.105350] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Smith KB, Murack M, Ismail N. The sex-dependent and enduring impact of pubertal stress on health and disease. Brain Res Bull 2023; 200:110701. [PMID: 37422090 DOI: 10.1016/j.brainresbull.2023.110701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/02/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Illness is often predicated long before the manifestation of its symptoms. Exposure to stressful experiences particularly during critical periods of development, such as puberty and adolescence, can induce various physical and mental illnesses. Puberty is a critical period of maturation for neuroendocrine systems, such as the hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-adrenal (HPA) axes. Exposure to adverse experiences during puberty can impede normal brain reorganizing and remodelling and result in enduring consequences on brain functioning and behaviour. Stress responsivity differs between the sexes during the pubertal period. This sex difference is partly due to differences in circulating sex hormones between males and females, impacting stress and immune responses differently. The effects of stress during puberty on physical and mental health remains under-examined. The purpose of this review is to summarize the most recent findings pertaining to age and sex differences in HPA axis, HPG axis, and immune system development, and describe how disruption in the functioning of these systems can propagate disease. Lastly, we delve into the notable neuroimmune contributions, sex differences, and the mediating role of the gut microbiome on stress and health outcomes. Understanding the enduring consequences of adverse experiences during puberty on physical and mental health will allow a greater proficiency in treating and preventing stress-related diseases early in development.
Collapse
Affiliation(s)
- Kevin B Smith
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Michael Murack
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada; LIFE Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
6
|
Hashimoto JG, Singer ML, Goeke CM, Zhang F, Song Y, Xia K, Linhardt RJ, Guizzetti M. Sex differences in hippocampal structural plasticity and glycosaminoglycan disaccharide levels after neonatal handling. Exp Neurol 2023; 361:114313. [PMID: 36572372 PMCID: PMC10097408 DOI: 10.1016/j.expneurol.2022.114313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
In this study we investigated the effects of a neonatal handling protocol that mimics the handling of sham control pups in protocols of neonatal exposure to brain insults on dendritic arborization and glycosaminoglycan (GAG) levels in the developing brain. GAGs are long, unbranched polysaccharides, consisting of repeating disaccharide units that can be modified by sulfation at specific sites and are involved in modulating neuronal plasticity during brain development. In this study, male and female Sprague-Dawley rats underwent neonatal handling daily between post-natal day (PD)4 and PD9, with brains analyzed on PD9. Neuronal morphology and morphometric analysis of the apical and basal dendritic trees of CA1 hippocampal pyramidal neurons were carried out by Golgi-Cox staining followed by neuron tracing and analysis with the software Neurolucida. Chondroitin sulfate (CS)-, Hyaluronic Acid (HA)-, and Heparan Sulfate (HS)-GAG disaccharide levels were quantified in the hippocampus by Liquid Chromatography/Mass Spectrometry analyses. We found sex by neonatal handling interactions on several parameters of CA1 pyramidal neuron morphology and in the levels of HS-GAGs, with females, but not males, showing an increase in both dendritic arborization and HS-GAG levels. We also observed increased expression of glucocorticoid receptor gene Nr3c1 in the hippocampus of both males and females following neonatal handling suggesting that both sexes experienced a similar stress during the handling procedure. This is the first study to show sex differences in two parameters of brain plasticity, CA1 neuron morphology and HS-GAG levels, following handling stress in neonatal rats.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Mo L Singer
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Calla M Goeke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Yuefan Song
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Ke Xia
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
7
|
Sex differences in addiction-relevant behavioral outcomes in rodents following early life stress. ADDICTION NEUROSCIENCE 2023; 6. [PMID: 37101684 PMCID: PMC10124992 DOI: 10.1016/j.addicn.2023.100067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In humans, exposure to early life stress (ELS) is an established risk factor for the development of substance use disorders (SUDs) during later life. Similarly, rodents exposed to ELS involving disrupted mother-infant interactions, such as maternal separation (MS) or adverse caregiving due to scarcity-adversity induced by limited bedding and nesting (LBN) conditions, also exhibit long-term alterations in alcohol and drug consumption. In both humans and rodents, there is a range of addiction-related behaviors that are associated with drug use and even predictive of subsequent SUDs. In rodents, these include increased anxiety-like behavior, impulsivity, and novelty-seeking, altered alcohol and drug intake patterns, as well as disrupted reward-related processes involving consummatory and social behaviors. Importantly, the expression of these behaviors often varies throughout the lifespan. Moreover, preclinical studies suggest that sex differences play a role in how exposure to ELS impacts reward and addiction-related phenotypes as well as underlying brain reward circuitry. Here, addiction-relevant behavioral outcomes and mesolimbic dopamine (DA) dysfunction resulting from ELS in the form of MS and LBN are discussed with a focus on age- and sex-dependent effects. Overall, these findings suggest that ELS may increase susceptibility for later life drug use and SUDs by interfering with the normal maturation of reward-related brain and behavioral function.
Collapse
|
8
|
Bucknor MC, Gururajan A, Dale RC, Hofer MJ. A comprehensive approach to modeling maternal immune activation in rodents. Front Neurosci 2022; 16:1071976. [PMID: 36590294 PMCID: PMC9800799 DOI: 10.3389/fnins.2022.1071976] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Prenatal brain development is a highly orchestrated process, making it a very vulnerable window to perturbations. Maternal stress and subsequent inflammation during pregnancy leads to a state referred to as, maternal immune activation (MIA). If persistent, MIA can pose as a significant risk factor for the manifestation of neurodevelopmental disorders (NDDs) such as autism spectrum disorder and schizophrenia. To further elucidate this association between MIA and NDD risk, rodent models have been used extensively across laboratories for many years. However, there are few uniform approaches for rodent MIA models which make not only comparisons between studies difficult, but some established approaches come with limitations that can affect experimental outcomes. Here, we provide researchers with a comprehensive review of common experimental variables and potential limitations that should be considered when designing an MIA study based in a rodent model. Experimental variables discussed include: innate immune stimulation using poly I:C and LPS, environmental gestational stress paradigms, rodent diet composition and sterilization, rodent strain, neonatal handling, and the inclusion of sex-specific MIA offspring analyses. We discuss how some aspects of these variables have potential to make a profound impact on MIA data interpretation and reproducibility.
Collapse
Affiliation(s)
- Morgan C. Bucknor
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Anand Gururajan
- The Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Russell C. Dale
- The Children’s Hospital at Westmead, Kids Neuroscience Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Markus J. Hofer
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Roque A, Valles Méndez KM, Ruiz R, Pineda E, Lajud N. Early life stress induces a transient increase in hippocampal corticotropin-releasing hormone in rat neonates that precedes the effects on hypothalamic neuropeptides. Eur J Neurosci 2022; 55:2108-2121. [PMID: 33745155 DOI: 10.1111/ejn.15193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Early life stress (ELS) programs hypothalamus-pituitary-adrenal (HPA) axis activity and affects synaptic plasticity and cognitive performance in adults; however, the effects of ELS during the temporal window of vulnerability are poorly understood. This study aimed to thoroughly characterize the effects of ELS in the form of periodic maternal separation (MS180) during the time of exposure to stress. Hippocampal corticotropin-releasing hormone (CRH) gene expression and baseline HPA axis activity were analyzed at postnatal (P) days 6, 12, 15, and 21, and in adulthood (P75); these factors were correlated with plasticity markers and adult behavior. Our results indicate that MS180 induces an increase in hippocampal CRH expression at P9, P12, and P15, whereas an increase in hypothalamic CRH expression was observed from P12 to P21. Increased arginine-vasopressin expression and corticosterone levels were observed only at P21. Moreover, MS180 caused transient alterations in hypothalamic synaptophysin expression during early life. As adults, MS180 rats showed a passive coping strategy in the forced swimming test, cognitive impairments in the object location test, increased hypothalamic CRH expression, and decreased oxytocin (OXT) expression. Spearman's analysis indicated that cognitive impairments correlated with CRH and OXT expression. In conclusion, our data indicate that MS180 induces a transient increase in hippocampal CRH expression in neonates that precedes the effects on hypothalamic neuropeptides, confirming the role of increased CRH during the temporal window of vulnerability as a mediator of some of the detrimental effects of ELS on brain development and adult behavior.
Collapse
Affiliation(s)
- Angélica Roque
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Kinberli Marcela Valles Méndez
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Roberto Ruiz
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Edel Pineda
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Naima Lajud
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| |
Collapse
|
10
|
Jethava V, Kadish J, Kakonge L, Wiseman-Hakes C. Early Attachment and the Development of Social Communication: A Neuropsychological Approach. Front Psychiatry 2022; 13:838950. [PMID: 35463524 PMCID: PMC9024310 DOI: 10.3389/fpsyt.2022.838950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Social communication forms the foundation of human relationships. Social communication, i.e., the appropriate understanding and use of verbal and non-verbal communication within a social context, profoundly impacts mental health across the lifespan and is also highly vulnerable to neurodevelopmental threats and social adversities. There exists a strong interconnection between the development of language and other higher cognitive skills, mediated, in part, through the early attachment relationship. Consideration of how attachment links to brain development can help us understand individuals with social communication difficulties across the lifespan. The early attachment relationship supports the development of the foundational constructs of social communication. In this paper, a neuropsychological perspective was applied to social communication, which integrated evidence from early attachment theory, examining the underpinnings of social communication components identified by the SoCom model, namely socio-cognitive, socio-emotional, and socio-linguistic constructs. A neuropsychological perspective underscores the importance of interdisciplinary collaboration. This should also inform approaches to prevention, policy, intervention, and advocacy for individuals with or at risk for social communication impairments, as well as their families.
Collapse
Affiliation(s)
- Vibhuti Jethava
- York Hills Centre for Children, Youth and Families, Richmond Hill, ON, Canada
- Department of Speech-Language Pathology, University of Toronto, Toronto, ON, Canada
| | - Jocelyn Kadish
- Department of Speech-Language Pathology, University of Toronto, Toronto, ON, Canada
| | - Lisa Kakonge
- Department of Speech-Language Pathology, University of Toronto, Toronto, ON, Canada
- School of Rehabilitation Science, Speech Language Pathology Program, McMaster University, Hamilton, ON, Canada
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Catherine Wiseman-Hakes
- School of Rehabilitation Science, Speech Language Pathology Program, McMaster University, Hamilton, ON, Canada
- KITE Research Institute, Toronto Rehab-University Health Network, Toronto, ON, Canada
| |
Collapse
|
11
|
Male long-Evans rats: An outbred model of marked hypothalamic-pituitary-adrenal hyperactivity. Neurobiol Stress 2021; 15:100355. [PMID: 34307794 PMCID: PMC8283147 DOI: 10.1016/j.ynstr.2021.100355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/08/2021] [Accepted: 06/13/2021] [Indexed: 11/21/2022] Open
Abstract
Rat and mouse strains differ in behavioral and physiological characteristics, and such differences can contribute to explain discrepant results between laboratories and better select the most appropriate strain for a particular purpose. Differences in the activity of the hypothalamic-pituitary-adrenal (HPA) axis are particularly important given the pivotal role of this system in determining consequences of exposure to stressors. In this regard, Long-Evans (LE) rats are widely used in stress research, but there is no specific study aiming at thoroughly characterizing HPA activity in LE versus other extensively used strains. In a first experiment, LE showed higher resting ACTH and corticosterone levels only at certain points of the circadian rhythm, but much greater ACTH responsiveness to stressors (novel environment and forced swim) than Sprague-Dawley (SD) rats. Accordingly, enhanced corticotropin-releasing hormone (CRH) expression in the paraventricular nucleus of the hypothalamus and reduced expression of glucocorticoid receptors were observed in the hippocampal formation. Additionally, they are hyperactive in novel environments, and prone to adopt passive-like behavior when compared to SD rats. Supporting that altered HPA function has a marked physiological impact, we observed in another set of animals much lower thymus weight in LE than SD rats. Finally, to demonstrate that LE rats are likely to have higher HPA responsiveness to stressors than most strains, we studied resting and stress levels of HPA hormones in LE versus Wistar and Fischer rats, the latter considered an example of high HPA responsiveness. Again, LE showed higher resting and stress levels of ACTH than both Wistar and Fischer rats. As ACTH responsiveness to stressors in LE rats is stronger than that previously reported when comparing other rat strains and they are commercially available, they could be an appropriate model for studying the behavioral and physiological implications of a hyper-active HPA axis under normal and pathological conditions. Strain differences in hypothalamic-pituitary-adrenal (HPA) function were studied. Long-Evans (LE) rats show greater HPA response to stressors than other strains. CRH expression in critical brain areas is greater in LE than Sprague-Dawley (SD) rats. Glucocorticoid receptor expression was lower in the hippocampal formation of LE rats. LE rats are more active in novel environments but showed more passive coping.
Collapse
|
12
|
Lajud N, Roque A, Cheng JP, Bondi CO, Kline AE. Early Life Stress Preceding Mild Pediatric Traumatic Brain Injury Increases Neuroinflammation but Does Not Exacerbate Impairment of Cognitive Flexibility during Adolescence. J Neurotrauma 2020; 38:411-421. [PMID: 33040677 DOI: 10.1089/neu.2020.7354] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Early life stress (ELS) followed by pediatric mild traumatic brain injury (mTBI) negatively impacts spatial learning and memory and increases microglial activation in adolescent rats, but whether the same paradigm negatively affects higher order executive function is not known. Hence, we utilized the attentional set-shifting test (AST) to evaluate executive function (cognitive flexibility) and to determine its relationship with neuroinflammation and hypothalamic-pituitary-adrenal (HPA) axis activity after pediatric mTBI in male rats. ELS was induced via maternal separation for 180 min per day (MS180) during the first 21 post-natal (P) days, while controls (CONT) were undisturbed. At P21, fully anesthetized rats received a mild controlled cortical impact (2.2 mm tissue deformation at 4 m/sec) or sham injury. AST was evaluated during adolescence on P35-P40 and cytokine expression and HPA activity were analyzed on P42. The data indicate that pediatric mTBI produced a significant reversal learning deficit on the AST versus sham (p < 0.05), but that the impairment was not exacerbated further by MS180. Additionally, ELS produced an overall elevation in set-loss errors on the AST, and increased hippocampal interleukin (IL)-1β expression after TBI. A significant correlation was observed in executive dysfunction and IL-1β expression in the ipsilateral pre-frontal cortex and hippocampus. Although the combination of ELS and pediatric mTBI did not worsen executive function beyond that of mTBI alone (p > 0.05), it did result in increased hippocampal neuroinflammation relative to mTBI (p < 0.05). These findings provide important insight into the susceptibility to incur alterations in cognitive and neuroimmune functioning after stress exposure and TBI during early life.
Collapse
Affiliation(s)
- Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, México
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, México.,Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey P Cheng
- Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Corina O Bondi
- Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony E Kline
- Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Diaz-Chávez A, Lajud N, Roque A, Cheng JP, Meléndez-Herrera E, Valdéz-Alarcón JJ, Bondi CO, Kline AE. Early life stress increases vulnerability to the sequelae of pediatric mild traumatic brain injury. Exp Neurol 2020; 329:113318. [PMID: 32305419 DOI: 10.1016/j.expneurol.2020.113318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Early life stress (ELS) is a risk factor for many psychopathologies that happen later in life. Although stress can occur in cases of child abuse, studies on non-accidental brain injuries in pediatric populations do not consider the possible increase in vulnerability caused by ELS. Hence, we sought to determine whether ELS increases the effects of pediatric mild traumatic brain injury (mTBI) on cognition, hippocampal inflammation, and plasticity. Male rats were subjected to maternal separation for 180 min per day (MS180) or used as controls (CONT) during the first 21 post-natal (P) days. At P21 the rats were anesthetized with isoflurane and subjected to a mild controlled cortical impact or sham injury. At P32 the rats were injected with the cell proliferation marker bromodeoxyuridine (BrdU, 500 mg/kg), then evaluated for spatial learning and memory in a water maze (P35-40) and sacrificed for quantification of Ki67+, BrdU+ and Iba1+ (P42). Neither MS180 nor mTBI impacted cognitive outcome when provided alone but their combination (MS180 + mTBI) decreased spatial learning and memory relative to Sham controls (p < .01). mTBI increased microglial activation and affected BrdU+ cell survival in the ipsilateral hippocampus without affecting proliferation rates. However, only MS180 + mTBI increased microglial activation in the area adjacent to the injury and the contralateral CA1 hippocampal subfield, and decreased cell proliferation in the ipsilateral neurogenic niche. Overall, the data show that ELS increases the vulnerability to the sequelae of pediatric mTBI and may be mediated by increased neuroinflammation.
Collapse
Affiliation(s)
- Arturo Diaz-Chávez
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Juan José Valdéz-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Psychology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
14
|
Cirulli F, Musillo C, Berry A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020; 447:122-135. [PMID: 32032668 DOI: 10.1016/j.neuroscience.2020.01.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
Maternal obesity plays a key role in the health trajectory of the offspring. Although research on this topic has largely focused on the potential of this condition to increase the risk for child obesity, it is becoming more and more evident that it can also significantly impact cognitive function and mental health. The mechanisms underlying these effects are starting to be elucidated and point to the placenta as a critical organ that may mediate changes in the response to stress, immune function and oxidative stress. Long-term effects of maternal obesity may rely upon epigenetic changes in selected genes that are involved in metabolic and trophic regulations of the brain. More recent evidence also indicates the gut microbiota as a potential mediator of these effects. Overall, understanding cause-effect relationships can allow the development of preventive measures that could rely upon dietary changes in the mother and the offspring. Addressing diets appears more feasible than developing new pharmacological targets and has the potential to affect the multiple interconnected physiological pathways engaged by these complex regulations, allowing prevention of both metabolic and mental disorders.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
15
|
Yang R, Sun H, Wu Y, Lu G, Wang Y, Li Q, Zhou J, Sun H, Sun L. Long-Lasting Sex-Specific Effects Based On Emotion- and Cognition-Related Behavioral Assessment of Adult Rats After Post-Traumatic Stress Disorder From Different Lengths of Maternal Separation. Front Psychiatry 2019; 10:289. [PMID: 31231246 PMCID: PMC6558979 DOI: 10.3389/fpsyt.2019.00289] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Adverse early life stress is a major cause of vulnerability to various mental disorders in adulthood, including post-traumatic stress disorder (PTSD). Recent studies have suggested that early life stress can help the body adapt optimally when faced with stressful trauma in adult life. An interaction may exist between early life stress (e.g., childhood trauma) and vulnerability to PTSD. This study aimed to evaluate emotion-related behaviors and verify the long-lasting effects of cognitive aspects of PTSD after exposure to severe adverse early life stress, such as long-term separation. Adverse early life stress was simulated by subjecting rats to 3 or 6 consecutive hours of maternal separation (MS) daily, from postnatal day (PND) 2 to PND 14. Single-prolonged stress (SPS) was simulated on PND 80 to imitate other adulthood stresses of PTSD with gender divisions (M-MS3h-PTSD, F-MS3h-PTSD, M-MS6h-PTSD, F-MS6h-PTSD, M-PTSD, and F-PTSD). After the MS and PTSD sessions, behavioral tests were conducted to assess the effectiveness of these treatments, which included an open field test (OFT), elevated plus maze test (EPMT), water maze test (WMT), and forced swimming test (FST) to detect anxiety-like behavior (OFT and EPMT), memory behavior (WMT), and depressive behavior (FST). The M-MS3h-PTSD group had fewer time entries into the open arms of EPMT than the F-MS3h-PTSD group, and the M-MS6h-PTSD group demonstrated fewer up-right postures in the OFT than the F-MS6h-PTSD group. The M-MS3h-PTSD group exhibited more exploratory behavior than the M-MS6h-PTSD and M-PTSD groups in the OFT. Less exploratory behavior was observed in the F-MS3h-PTSD group than in the F-MS6h-PTSD group, which demonstrated significantly increased freezing times in the FST compared to the F-PTSD group. The WMT revealed significant differences in learning and memory performance between the M-MS3h-PTSD group and other treatment groups, which were not found in the female rats. These findings demonstrate that an early stressful experience, such as MS, may be involved in helping the body adapt optimally when faced with additional trauma in adulthood, although mild early life stress might benefit learning and memory among males.
Collapse
Affiliation(s)
- Rucui Yang
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Haoran Sun
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yani Wu
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Guohua Lu
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Yanyu Wang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, Hong Kong
| | - Jin Zhou
- College of Pharmacy, Weifang Medical University, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Lin Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| |
Collapse
|
16
|
Sagar RL, Cockrem J, Rayner MJ, Stanley MC, Welch J, Dunphy BJ. Regular handling reduces corticosterone stress responses to handling but not condition of semi-precocial mottled petrel (Pterodroma inexpectata) chicks. Gen Comp Endocrinol 2019; 272:1-8. [PMID: 30419229 DOI: 10.1016/j.ygcen.2018.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 10/24/2018] [Accepted: 11/04/2018] [Indexed: 10/27/2022]
Abstract
Handling of avian study species is common in ecological research, yet few studies account for the impact of handling in nestlings where exposure to stress may result in negative lifetime fitness consequences. As a result, our understanding of stress reactivity in free-living avian young is limited. In this study we examined the cumulative impact of three levels of research-relevant handling (control, daily and every three days) on the development of the stress response, growth and condition of semi-precocial seabird chicks from near-hatching to near-fledging. By measuring corticosterone concentrations in plasma, we found that mottled petrel (Pterodroma inexpectata) chicks were capable of mounting a stress response comparable to adults from near-hatching. There were no differences in plasma corticosterone concentrations in initial samples (<4 min) between groups at six weeks of age, though by 12 weeks of age plasma corticosterone concentrations in initial samples collected from chicks handled daily were lower than chicks that were handled once every three days, and from control chicks. Corticosterone responses to handling were lower in chicks handled daily at six and 12 weeks of age when compared to other handling groups. Handling chicks daily or every three days had no negative effect on the growth or condition of chicks when compared to control chicks. These findings indicate that daily handling results in chicks became accustomed to handling, with no evidence that regular handling was detrimental to mottled petrel chicks. However, given the unique life-history characteristics of mottled petrels relative to closely related species, we caution that this finding may be species-specific, and wider testing is recommended.
Collapse
Affiliation(s)
- Rachael L Sagar
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - John Cockrem
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North 4442, New Zealand
| | - Matt J Rayner
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Auckland War Memorial Museum, The Domain, Private Bag 92018, Victoria St West, Auckland 1142, New Zealand
| | - Margaret C Stanley
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jemma Welch
- Department of Conservation, Wairepo Road, Twizel 7901, New Zealand
| | - Brendon J Dunphy
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
17
|
Liu PZ, Nusslock R. How Stress Gets Under the Skin: Early Life Adversity and Glucocorticoid Receptor Epigenetic Regulation. Curr Genomics 2018; 19:653-664. [PMID: 30532645 PMCID: PMC6225447 DOI: 10.2174/1389202919666171228164350] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 05/31/2017] [Accepted: 12/17/2017] [Indexed: 11/22/2022] Open
Abstract
Early life adversity is associated with both persistent disruptions in the hypothalamic-pituitary-adrenal (HPA) axis and psychiatric symptoms. Glucocorticoid receptors (GRs), which are encoded by the NR3C1 gene, bind to cortisol and other glucocorticoids to create a negative feedback loop within the HPA axis to regulate the body's neuroendocrine response to stress. Excess methylation of a promoter sequence within NR3C1 that attenuates GR expression, however, has been associated with both early life adversity and psychopathology. As critical regulators within the HPA axis, GRs and their epigenetic regulation may mediate the link between early life adversity and the onset of psychopathology. The present review discusses this work as one mechanism by which stress may get under the skin to disrupt HPA functioning at an epigenetic level and create long-lasting vulnerabilities in the stress regulatory system that subsequently predispose individuals to psychopathology. Spanning prenatal influences to critical periods of early life and adolescence, we detail the impact that early adversity has on GR expression, physiological responses to stress, and their implications for long-term stress management. We next propose a dual transmission hypothesis regarding both genomic and non-genomic mechanisms by which chronic and acute stress propagate through numerous generations. Lastly, we outline several directions for future research, including potential reversibility of methylation patterns and its functional implications, variation in behavior determined solely by NR3C1, and consensus on which specific promoter regions should be studied.
Collapse
Affiliation(s)
- Patrick Z. Liu
- Department of Psychology, Northwestern University, Evanston, IL60208, USA
| | - Robin Nusslock
- Department of Psychology, Northwestern University, Evanston, IL60208, USA
| |
Collapse
|
18
|
Miragaia AS, de Oliveira Wertheimer GS, Consoli AC, Cabbia R, Longo BM, Girardi CEN, Suchecki D. Maternal Deprivation Increases Anxiety- and Depressive-Like Behaviors in an Age-Dependent Fashion and Reduces Neuropeptide Y Expression in the Amygdala and Hippocampus of Male and Female Young Adult Rats. Front Behav Neurosci 2018; 12:159. [PMID: 30131681 PMCID: PMC6090069 DOI: 10.3389/fnbeh.2018.00159] [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: 05/05/2018] [Accepted: 07/09/2018] [Indexed: 01/28/2023] Open
Abstract
Maternal deprivation for 24 h produces an immediate increase in basal and stress-induced corticosterone (CORT) secretion. Given the impact of elevated CORT levels on brain development, the goal of the present study was to characterize the effects of maternal deprivation at postnatal days 3 (DEP3) or 11 (DEP11) on emotional behavior and neuropeptide Y immunoreactivity (NPY-ir) in the basolateral amygdala (BLA) and dorsal hippocampus (dHPC) of male and female rats. Litters were distributed in control non-deprived (CTL), DEP3, or DEP11 groups. In Experiment 1, within each litter, one male and one female were submitted to one of the following tests: novelty suppressed feeding (NSF), sucrose negative contrast test (SNCT), and forced swimming test (FST), between postnatal days 52 and 60. In Experiment 2, two males and two females per litter were exposed to the elevated plus maze and 1 h later, perfused for investigation of NPY-ir, on PND 52. The results showed that DEP3 rats displayed greater anxiety-like behavior in the NSF and EPM, compared to CTL and DEP11 counterparts. In the SNCT, DEP3 and DEP11 males showed less suppression of the lower sucrose concentration intake, whereas all females suppressed less than males. Both manipulated groups displayed more immobility in the FST, although this effect was greater in DEP3 than in DEP11 rats. NPY-ir was reduced in DEP3 and DEP11 males and females in the BLA, whereas in the dHPC, DEP3 males showed less NPY-ir than DEP11, which, in turn, presented less NPY-ir than CTL rats. Females showed less NPY-ir than males in both structures. Because the deprivation effects were more intense in DEP3 than in DEP11, in Experiment 3, the frequency of nursing posture, licking-grooming, and interaction with pups was assessed upon litter reunion with mothers. Mothers of DEP11 litters engaged more in anogenital licking than mothers of DEP3 litters. The present results indicate that maternal deprivation changed affective behavior with greater impact in the earlier age and reduced the expression of NPY in emotion-related brain areas. The age-dependent differential effects of deprivation on maternal behavior could, at least in part, explain the outcomes in young adult rats.
Collapse
Affiliation(s)
- Alexandra S Miragaia
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Amanda C Consoli
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rafael Cabbia
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz M Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carlos E N Girardi
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Lupien SJ, Juster RP, Raymond C, Marin MF. The effects of chronic stress on the human brain: From neurotoxicity, to vulnerability, to opportunity. Front Neuroendocrinol 2018; 49:91-105. [PMID: 29421159 DOI: 10.1016/j.yfrne.2018.02.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 01/12/2023]
Abstract
For the last five decades, science has managed to delineate the mechanisms by which stress hormones can impact on the human brain. Receptors for glucocorticoids are found in the hippocampus, amygdala and frontal cortex, three brain regions involved in memory processing and emotional regulation. Studies have shown that chronic exposure to stress is associated with reduced volume of the hippocampus and that chronic stress can modulate volumes of both the amygdala and frontal cortex, suggesting neurotoxic effects of stress hormones on the brain. Yet, other studies report that exposure to early adversity and/or familial/social stressors can increase vulnerability to stress in adulthood. Models have been recently developed to describe the roles that neurotoxic and vulnerability effects can have on the developing brain. These models suggest that developing early stress interventions could potentially counteract the effects of chronic stress on the brain and results going along with this hypothesis are summarized.
Collapse
Affiliation(s)
- Sonia J Lupien
- Centre for Studies on Human Stress, Montreal Mental Health University Institute, Canada; Department of Psychiatry, Faculty of Medicine, University of Montreal, Canada.
| | - Robert-Paul Juster
- Centre for Studies on Human Stress, Montreal Mental Health University Institute, Canada; Department of Psychiatry, Columbia University, New York, United States
| | - Catherine Raymond
- Centre for Studies on Human Stress, Montreal Mental Health University Institute, Canada; Department of Neurosciences, Université de Montreal, Canada
| | - Marie-France Marin
- Centre for Studies on Human Stress, Montreal Mental Health University Institute, Canada; Department of Psychiatry, Faculty of Medicine, University of Montreal, Canada
| |
Collapse
|
20
|
Lalonde R, Strazielle C. Neuroanatomical pathways underlying the effects of hypothalamo-hypophysial-adrenal hormones on exploratory activity. Rev Neurosci 2018; 28:617-648. [PMID: 28609296 DOI: 10.1515/revneuro-2016-0075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/16/2017] [Indexed: 12/25/2022]
Abstract
When injected via the intracerebroventricular route, corticosterone-releasing hormone (CRH) reduced exploration in the elevated plus-maze, the center region of the open-field, and the large chamber in the defensive withdrawal test. The anxiogenic action of CRH in the elevated plus-maze also occurred when infused in the basolateral amygdala, ventral hippocampus, lateral septum, bed nucleus of the stria terminalis, nucleus accumbens, periaqueductal grey, and medial frontal cortex. The anxiogenic action of CRH in the defensive withdrawal test was reproduced when injected in the locus coeruleus, while the amygdala, hippocampus, lateral septum, nucleus accumbens, and lateral globus pallidus contribute to center zone exploration in the open-field. In addition to elevated plus-maze and open-field tests, the amygdala appears as a target region for CRH-mediated anxiety in the elevated T-maze. Thus, the amygdala is the principal brain region identified with these three tests, and further research must identify the neural circuits underlying this form of anxiety.
Collapse
Affiliation(s)
| | - Catherine Strazielle
- , Laboratoire 'Stress, Immunité, Pathogènes' EA 7300 and Service de Microscopie Electronique, Faculté de Médecine
| |
Collapse
|
21
|
Raciti M, Ceccatelli S. Epigenetic mechanisms in developmental neurotoxicity. Neurotoxicol Teratol 2018; 66:94-101. [DOI: 10.1016/j.ntt.2017.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/28/2022]
|
22
|
Johnson FK, Kaffman A. Early life stress perturbs the function of microglia in the developing rodent brain: New insights and future challenges. Brain Behav Immun 2018; 69:18-27. [PMID: 28625767 PMCID: PMC5732099 DOI: 10.1016/j.bbi.2017.06.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/21/2017] [Accepted: 06/14/2017] [Indexed: 11/24/2022] Open
Abstract
The role of the innate immune system in mediating some of the consequences of childhood abuse and neglect has received increasing attention in recent years. Most of the work to date has focused on the role that neuroinflammation plays in the long-term adult psychiatric and medical complications associated with childhood maltreatment. The effects of stress-induced neuroinflammation on neurodevelopment have received little attention because until recently this issue has not been studied systematically in animal models of early life stress. The primary goal of this review is to explore the hypothesis that elevated corticosterone during the first weeks of life in mice exposed to brief daily separation (BDS), which is a mouse model of early life stress, disrupts microglial function during a critical period of brain development. We propose that perturbations of microglial function lead to abnormal maturation of several neuronal and non-neuronal cellular processes resulting in behavioral abnormalities that emerge during the juvenile period and persist in adulthood. Here, we highlight recent work demonstrating that exposure to BDS alters microglial cell number, morphology, phagocytic activity, and gene expression in the developing hippocampus in a manner that extends into the juvenile period. These changes in microglial function are associated with abnormalities in developmental processes mediated by microglia including synaptogenesis, synaptic pruning, axonal growth, and myelination. We examine the changes in microglial gene expression in the context of previous work demonstrating developmental and behavioral abnormalities in BDS mice and in other animal models of early life stress. The possible utility of these findings for developing novel PET imaging to assess microglial function in individuals exposed to childhood maltreatment is also discussed.
Collapse
Affiliation(s)
- Frances K Johnson
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| |
Collapse
|
23
|
Karbaschi R, Zardooz H, Khodagholi F, Dargahi L, Salimi M, Rashidi F. Maternal high-fat diet intensifies the metabolic response to stress in male rat offspring. Nutr Metab (Lond) 2017; 14:20. [PMID: 28261314 PMCID: PMC5329934 DOI: 10.1186/s12986-017-0177-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mother's consumption of high-fat food can affect glucose metabolism and the hypothalamic-pituitary-adrenal axis responsiveness in the offspring and potentially affect the metabolic responses to stress as well. This study examines the effect of maternal high-fat diet on the expression of pancreatic glucose transporter 2 and the secretion of insulin in response to stress in offspring. METHODS Female rats were randomly divided into normal and high-fat diet groups and were fed in accordance with their given diets from pre-pregnancy to the end of lactation. The offspring were divided into control (NC and HFC) and stress (NS and HFS) groups based on their mothers' diet and exposure to stress in adulthood. After the two-week stress induction period was over, an intraperitoneal glucose tolerance test (IPGTT) was performed and plasma glucose and insulin levels were assessed. The pancreas was then removed for measuring insulin secretion from the isolated islets as well as glucose transporter 2 mRNA expression and protein levels. RESULTS According to the results obtained, plasma corticosterone concentrations increased significantly on days 1 and 14 of the stress induction period and were lower on the last day compared to on the first day. In both the NS and HFS groups, stress reduced plasma insulin concentration in the IPGTT without changing the plasma glucose concentration, suggesting an increased insulin sensitivity in the NS and HFS groups, although more markedly in the latter. Stress reduced insulin secretion (at high glucose concentrations) and increased glucose transporter 2 mRNA and protein expression, especially in the HFS group. CONCLUSION Mothers' high-fat diet appears to intensify the stress response by changing the programming of the neuroendocrine system in the offspring.
Collapse
Affiliation(s)
- Roxana Karbaschi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - FatemehSadat Rashidi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Pratt M, Goldstein A, Levy J, Feldman R. Maternal Depression Across the First Years of Life Impacts the Neural Basis of Empathy in Preadolescence. J Am Acad Child Adolesc Psychiatry 2017; 56:20-29.e3. [PMID: 27993224 DOI: 10.1016/j.jaac.2016.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/04/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Exposure to maternal depression across the first years of life markedly increases children's susceptibility to psychopathology, yet no study has tested its effects on the maturation of children's social brain. METHOD Using a birth cohort of mothers with no contextual risk (N = 1,983), families were followed at 7 time points from birth to 11 years and repeatedly assessed for maternal depression across the first 6 years to form 2 cohorts: mothers continuously depressed from birth to 6 years and controls without depression. At 11 years of age, children's (n = 72; depressed, n = 27; nondepressed, n = 45) brain response to others' pain was measured by magnetoencephalography. RESULTS Preadolescents displayed a unique oscillatory pattern with higher alpha power to pain versus no pain expressing as alpha rebound, not alpha suppression, at a late time window (1,100-1,300 ms post-stimulus) in the supplementary motor area. This suggests that top-down processing in areas of the pain matrix can underpin the maturation of vicarious empathy. Children of mothers with depression showed enhanced alpha rebound to pain in the right posterior superior temporal gyrus, which was unrelated to emotion detection abilities, pointing to decreased late processing of others' overwhelming experiences in socio-cognitive areas. Alpha power in the posterior superior temporal gyrus was predicted by higher maternal intrusiveness and lower synchrony across early childhood. CONCLUSION These findings, from the first study to examine maternal depression and early caregiving as long-term predictors of children's neural empathic response, pinpoint a decrease in top-down socio-cognitive mechanisms as potential pathways for the cross-generational transfer of vulnerability from mothers with depression to their offspring and highlight the need for early interventions focused on enhancing maternal attunement.
Collapse
Affiliation(s)
| | - Abraham Goldstein
- Bar-Ilan University, Ramat-Gan, Israel; Gonda Brain Sciences Center, Bar-Ilan University
| | - Jonathan Levy
- Bar-Ilan University, Ramat-Gan, Israel; Gonda Brain Sciences Center, Bar-Ilan University
| | - Ruth Feldman
- Bar-Ilan University, Ramat-Gan, Israel; Gonda Brain Sciences Center, Bar-Ilan University.
| |
Collapse
|
25
|
Perkeybile AM, Bales KL. Intergenerational transmission of sociality: the role of parents in shaping social behavior in monogamous and non-monogamous species. J Exp Biol 2017; 220:114-123. [PMID: 28057834 PMCID: PMC5278619 DOI: 10.1242/jeb.142182] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Social bonds are necessary for many mammals to survive and reproduce successfully. These bonds (i.e. pair-bonds, friendships, filial bonds) are characterized by different periods of development, longevity and strength. Socially monogamous species display certain behaviors not seen in many other mammals, such as adult pair-bonding and male parenting. In our studies of prairie voles (Microtus ochrogaster) and titi monkeys (Callicebus cupreus), we have examined the neurohormonal basis of these bonds. Here, we discuss the evidence from voles that aspects of adolescent and adult social behavior are shaped by early experience, including changes to sensory systems and connections, neuropeptide systems such as oxytocin and vasopressin, and alterations in stress responses. We will compare this with what is known about these processes during development and adulthood in other mammalian species, both monogamous and non-monogamous, and how our current knowledge in voles can be used to understand the development of and variation in social bonds. Humans are endlessly fascinated by the variety of social relationships and family types displayed by animal species, including our own. Social relationships can be characterized by directionality (either uni- or bi-directional), longevity, developmental epoch (infant, juvenile or adult) and strength. Research on the neurobiology of social bonds in animals has focused primarily on 'socially monogamous' species, because of their long-term, strong adult affiliative bonds. In this Review, we attempt to understand how the ability and propensity to form these bonds (or lack thereof), as well as the display of social behaviors more generally, are transmitted both genomically and non-genomically via variation in parenting in monogamous and non-monogamous species.
Collapse
Affiliation(s)
- Allison M Perkeybile
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
- The Kinsey Institute, Indiana University, Bloomington, IN 47405, USA
| | - Karen L Bales
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
26
|
Delpech JC, Wei L, Hao J, Yu X, Madore C, Butovsky O, Kaffman A. Early life stress perturbs the maturation of microglia in the developing hippocampus. Brain Behav Immun 2016; 57:79-93. [PMID: 27301858 PMCID: PMC5010940 DOI: 10.1016/j.bbi.2016.06.006] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 12/22/2022] Open
Abstract
Children exposed to abuse or neglect show abnormal hippocampal development and similar findings have been reported in rodent models. Using brief daily separation (BDS), a mouse model of early life stress, we previously showed that exposure to BDS impairs hippocampal function in adulthood and perturbs synaptic maturation, synaptic pruning, axonal growth and myelination in the developing hippocampus. Given that microglia are involved in these developmental processes, we tested whether BDS impairs microglial activity in the hippocampus of 14 (during BDS) and 28-day old mice (one week after BDS). We found that BDS increased the density and altered the morphology of microglia in the hippocampus of 14-day old pups, effects that were no longer present on postnatal day (PND) 28. Despite the normal cell number and morphology seen at PND28, the molecular signature of hippocampal microglia, assessed using the NanoString immune panel, was altered at both ages. We showed that during normal hippocampal development, microglia undergo significant changes between PND14 and PND28, including reduced cell density, decreased ex vivo phagocytic activity, and an increase in the expression of genes involved in inflammation and cell migration. However, microglia harvested from the hippocampus of 28-day old BDS mice showed an increase in phagocytic activity and reduced expression of genes that normally increase across development. Promoter analysis indicated that alteration in the transcriptional activity of PU.1, Creb1, Sp1, and RelA accounted for most of the transcriptional changes seen during normal microglia development and for most of the BDS-induced changes at PND14 and PND28. These findings are the first to demonstrate that early life stress dysregulates microglial function in the developing hippocampus and to identify key transcription factors that are likely to mediate these changes.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Lan Wei
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Jin Hao
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Xiaoqing Yu
- W.M. Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, CT 06511, USA
| | - Charlotte Madore
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Oleg Butovsky
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| |
Collapse
|
27
|
Better Utilization of Mouse Models of Neurodegenerative Diseases in Preclinical Studies: From the Bench to the Clinic. Methods Mol Biol 2016; 1438:311-47. [PMID: 27150098 DOI: 10.1007/978-1-4939-3661-8_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The major symptom of Alzheimer's disease is dementia progressing with age. Its clinical diagnosis is preceded by a long prodromal period of brain pathology that encompasses both formation of extracellular amyloid and intraneuronal tau deposits in the brain and widespread neuronal death. At present, familial cases of dementia provide the most promising foundation for modeling neurodegenerative tauopathies, a group of heterogeneous disorders characterized by prominent intracellular accumulation of hyperphosphorylated tau protein. In this chapter, we describe major behavioral hallmarks of tauopathies, briefly outline the genetics underlying familial cases, and discuss the arising implications for modeling the disease in transgenic mouse systems. The selection of tests performed to evaluate the phenotype of a model should be guided by the key behavioral hallmarks that characterize human disorder and their homology to mouse cognitive systems. We attempt to provide general guidelines and establish criteria for modeling dementia in a mouse; however, interpretations of obtained results should avoid a reductionist "one gene, one disease" explanation of model characteristics. Rather, the focus should be directed to the question of how the mouse genome can cope with the over-expression of the protein coded by transgene(s). While each model is valuable within its own constraints and the experiments performed are guided by specific hypotheses, we seek to expand upon their methodology by offering guidance spanning from issues of mouse husbandry to choices of behavioral tests and routes of drug administration that might increase the external validity of studies and consequently optimize the translational aspect of preclinical research.
Collapse
|
28
|
Colson V, Valotaire C, Geffroy B, Kiilerich P. Egg Cortisol Exposure Enhances Fearfulness in Larvae and Juvenile Rainbow Trout. Ethology 2015. [DOI: 10.1111/eth.12437] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Violaine Colson
- INRA; UR1037; Fish Physiology and Genomics; F-35000 Rennes France
| | | | - Benjamin Geffroy
- INRA; UR1037; Fish Physiology and Genomics; F-35000 Rennes France
| | - Pia Kiilerich
- INRA; UR1037; Fish Physiology and Genomics; F-35000 Rennes France
| |
Collapse
|
29
|
Jellyman JK, Valenzuela OA, Fowden AL. HORSE SPECIES SYMPOSIUM: Glucocorticoid programming of hypothalamic-pituitary-adrenal axis and metabolic function: Animal studies from mouse to horse1,2. J Anim Sci 2015; 93:3245-60. [DOI: 10.2527/jas.2014-8612] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- J. K. Jellyman
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502
| | - O. A. Valenzuela
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - A. L. Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
30
|
Early rearing experience is associated with vasopressin immunoreactivity but not reactivity to an acute non-social stressor in the prairie vole. Physiol Behav 2015; 147:149-56. [PMID: 25890274 DOI: 10.1016/j.physbeh.2015.04.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/06/2015] [Accepted: 04/12/2015] [Indexed: 01/06/2023]
Abstract
The early life experiences of an organism have the potential to alter its developmental trajectories. Perhaps one of the most powerful influences during this period is the parent-offspring relationship. Previous work in several mammalian species has demonstrated that parental care in early life and specifically maternal behavior can influence several adult outcomes in offspring, including affiliative and aggressive behavior, parental behavior, hypothalamic-pituitary-adrenal (HPA) functioning and risk of psychopathology. We have previously demonstrated that naturally occurring variation in the type and amount of care given to offspring in a biparental species, the prairie vole (Microtus ochrogaster), is related to social, anxiety-like, aggressive behaviors as well as HPA response to chronic and acute social stressors. Here we aim to determine the effects of early biparental care on HPA functioning and the interaction between early care and later reactivity to a forced swim test, an acute non-social stressor. Behavior during the swim test as well as several indicators of HPA activity, including plasma corticosterone (CORT), corticotropin releasing hormone immunoreactivity (CRH-ir), and vasopressin immunoreactivity (AVP-ir) were measured. Results here indicate an effect of early experience on AVP-ir but not CRH-ir or plasma CORT. There were no differences in CORT levels between high-contact (HC) and low-contact (LC) males or females for either control animals or after a swim stressor. CRH-ir was higher in the central amygdala following a swim test but was not influenced by early care. However, AVP-ir was not influenced by exposure to a swim stressor but was affected by early parental care in a sex-dependent manner. Female HC offspring had increased AVP-ir in the SON while HC male offspring had decreased AVP-ir in the PVN compared to their LC counterparts. The differential response of CRH and AVP to early experience and later stress, and the lack of an interaction between early care rearing and later adult stress, suggest an independence in response of some components of the HPA system. In addition, these findings expand our understanding of the relationship between naturally occurring variation in early biparental care and sexual dimorphisms in adult outcomes.
Collapse
|
31
|
Sadeghimahalli F, Karbaschi R, Zardooz H, Khodagholi F, Rostamkhani F. Effect of early life stress on pancreatic isolated islets' insulin secretion in young adult male rats subjected to chronic stress. Endocrine 2015; 48:493-503. [PMID: 25030548 DOI: 10.1007/s12020-014-0337-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/10/2014] [Indexed: 01/12/2023]
Abstract
Early stressful experiences may predispose organisms to certain disorders, including those of metabolic defects. This study aimed to explore the effects of early life stress on pancreatic insulin secretion and glucose transporter 2 (GLUT2) protein levels in stressed young adult male rats. Foot shock stress was induced in early life (at 2 weeks of age) and/or in young adulthood (at 8-10 weeks of age) for five consecutive days. Blood samples were taken before and after stress exposure in young adult rats. At the end of the experiment, glucose tolerance, isolated islets' insulin secretion, and pancreatic amounts of GLUT2 protein were measured. Our results show that early life stress has no effect on basal plasma corticosterone levels and adrenal weight, either alone or combined with young adulthood stress, but that early life + young adulthood stress could prevent weight gain, and cause an increase in basal plasma glucose and insulin. The homeostasis model assessment of insulin resistance index did not increase, when the rats were subjected to early life stress alone, but increased when combined with young adulthood stress. Moreover, glucose tolerance was impaired by the combination of early life + young adult stress. There was a decrease in islet's insulin secretion in rats subjected to early life stress in response to 5.6 mM glucose concentration, but an increase with a concentration of 16.7 mM glucose. However, in rats subjected to early life + young adulthood stress, islet's insulin secretion increased in response to both the levels of glucose concentrations. GLUT2 protein levels decreased in response to early life stress and early life + young adulthood stress, but there was a greater decrease in the early life stress group. In conclusion, perhaps early life stress sensitizes the body to stressors later in life, making it more susceptible to metabolic syndrome only when the two are in combination.
Collapse
Affiliation(s)
- Forouzan Sadeghimahalli
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
32
|
Cooper MA, Clinard CT, Morrison KE. Neurobiological mechanisms supporting experience-dependent resistance to social stress. Neuroscience 2015; 291:1-14. [PMID: 25677096 PMCID: PMC4369454 DOI: 10.1016/j.neuroscience.2015.01.072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/28/2015] [Accepted: 01/30/2015] [Indexed: 11/16/2022]
Abstract
Humans and other animals show a remarkable capacity for resilience following traumatic, stressful events. Resilience is thought to be an active process related to coping with stress, although the cellular and molecular mechanisms that support active coping and stress resistance remain poorly understood. In this review, we focus on the neurobiological mechanisms by which environmental and social experiences promote stress resistance. In male Syrian hamsters, exposure to a brief social defeat stressor leads to increased avoidance of novel opponents, which we call conditioned defeat. Also, hamsters that have achieved dominant social status show reduced conditioned defeat as well as cellular and molecular changes in the neural circuits controlling the conditioned defeat response. We propose that experience-dependent neural plasticity occurs in the prelimbic (PL) cortex, infralimbic (IL) cortex, and ventral medial amygdala (vMeA) during the maintenance of dominance relationships, and that adaptations in these neural circuits support stress resistance in dominant individuals. Overall, behavioral treatments that promote success in competitive interactions may represent valuable interventions for instilling resilience.
Collapse
Affiliation(s)
- M A Cooper
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA.
| | - C T Clinard
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - K E Morrison
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
33
|
Perkeybile AM, Bales KL. Early rearing experience is related to altered aggression and vasopressin production following chronic social isolation in the prairie vole. Behav Brain Res 2015; 283:37-46. [PMID: 25623420 DOI: 10.1016/j.bbr.2015.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 01/07/2023]
Abstract
Parent-offspring interactions early in life can permanently shape the developmental path of those offspring. Manipulation of maternal care has long been used to alter the early-life environment of infants and impacts their later social behavior, aggression, and physiology. More recently, naturally occurring variation in maternal licking and grooming behavior has been shown to result in differences in social behavior and stress physiology in adult offspring. We have developed a model of natural variation in biparental care in the prairie vole (Microtus ochrogaster) and have demonstrated an association between the amount of early care received and later social behavior. In this study, we investigate the relationship between early life care and later aggression and neuroendocrine responses following chronic social isolation. Male and female offspring were reared by their high-contact (HC) or low-contact (LC) parents, then housed for 4 weeks post-weaning in social isolation. After 4 weeks, half of these offspring underwent an intrasexual aggression test. Brains and plasma were collected to measure corticotropin-releasing hormone (CRH) and vasopressin (AVP) immunoreactivity and plasma corticosterone (CORT). Male offspring of LC parents engaged in more aggressive behavior in the intrasexual aggression test compared to HC males. Female offspring of HC parents had higher plasma CORT levels after chronic social isolation and increases in the number and density of AVP-immunopositive cells in the supraoptic nucleus following an intrasexual aggression test. These findings show that the impact of early life biparental care on behavior and HPA activity following a social stressor is both sex-dependent and early experience-specific.
Collapse
Affiliation(s)
| | - Karen L Bales
- Department of Psychology, University of California-Davis, Davis, CA, USA
| |
Collapse
|
34
|
Dalmaz C, Noschang C, Krolow R, Raineki C, Lucion AB. How postnatal insults may program development: studies in animal models. ADVANCES IN NEUROBIOLOGY 2015; 10:121-47. [PMID: 25287539 DOI: 10.1007/978-1-4939-1372-5_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During the postnatal period, the nervous system is modified and shaped by experience, in order to adjust it to the particular environment in which the animal will live. This plasticity, one of the most remarkable characteristics of the nervous system, promotes adaptive changes, but it also makes brain more vulnerable to insults. This chapter will focus on the effects of interventions during the postnatal development in animal models of neonatal handling (usually up to 15 min of handling) and maternal separation (usually at least for 3 h). Sex-specific changes and effects of prepubertal stress such as social isolation later on in life were also considered. These interventions during development induce long-lasting traces in the pups' nervous system, which will be reflected in changes in neuroendocrine functions, including the hypothalamus-pituitary-adrenal and hypothalamus-pituitary-gonadal axes; anxiety and cognitive performance; and feeding, sexual, and social behavior. These enduring changes may be adaptive or maladaptive, depending on the environment in which the animal will live. The challenge researchers facing now is to determine how to reverse the deleterious effects that may result from early-life stress exposure.
Collapse
Affiliation(s)
- Carla Dalmaz
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porte Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
35
|
Abstract
Hormesis is the process by which small stresses build resilience to large stresses. We pre-exposed rats to various parameters of mild-to-moderate stress prior to traumatic stress in the present experiments to assess the potential benefits of hormetic training on resilience to traumatic, uncontrollable stress. Rats underwent varying stress pre-training parameters prior to exposure to uncontrollable traumatic stress in the learned helplessness procedure. The ability to prevent the exaggerated fear responding and escape deficits that normally follow experience with traumatic stress were used as a measure of the benefits of hormetic training. Four experiments examined the effects of number of training sessions, stressor severity and pattern of rest between pre-training stress sessions. Repeated exposure to mild restraint stress or moderate shock stress eliminated both the enhanced fear conditioning and shuttle-escape deficits that result from exposure to traumatic, inescapable shock. The pattern of rest did not contribute to resilience when the pre-exposure stressor was mild, but was vital when the pre-exposure stressor was moderate, with an alternation of stress and rest being the most effective procedure. The data also suggest that the level of resilience may increase with the number of pre-exposure sessions.
Collapse
Affiliation(s)
- Traci N Plumb
- Department of Psychology, University of California , Los Angeles, CA , USA
| | | | | |
Collapse
|
36
|
Abstract
Complex interactions between biological, behavioral and environmental factors are involved in mediating individual differences in health and disease. In this review, we present evidence suggesting that increased vulnerability to infectious disease may be at least, in part, due to long-lasting effects of early life psychosocial adversities. Studies have shown that maternal psychosocial stress during pregnancy is associated with long lasting changes in immune function and disease resistance in the offspring. Studies further indicated that harsh environmental conditions during the neonatal period may also cause lasting changes in host response to infectious disease. Although the mechanisms involved in these effects have not been fully examined, several potential mediators have been described, including changes in the development of the offspring hypothalamic-pituitary-adrenal axis, alterations in epigenetic pathways, stress-related maternal health risk behavior and infection during pregnancy. Although there are ample literature indicating that perinatal psychosocial stress increases vulnerability to disease, other reports suggest that mild predictable stressors may benefit the organism and allow better coping with future stressors. Thus, understanding the possible consequences of perinatal adversities and the mechanisms that are involved in immune regulation is important for increasing awareness to the potential outcomes of early negative life events and providing insight into potential therapies to combat infection in vulnerable individuals.
Collapse
Affiliation(s)
- Ronit Avitsur
- a School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo , Yaffo , Israel
| | - Sigal Levy
- a School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo , Yaffo , Israel
| | - Naama Goren
- a School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo , Yaffo , Israel
| | - Rachel Grinshpahet
- a School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo , Yaffo , Israel
| |
Collapse
|
37
|
Cañete T, Blázquez G, Tobeña A, Giménez-Llort L, Fernández-Teruel A. Cognitive and emotional alterations in young Alzheimer's disease (3xTgAD) mice: effects of neonatal handling stimulation and sexual dimorphism. Behav Brain Res 2014; 281:156-71. [PMID: 25446741 DOI: 10.1016/j.bbr.2014.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
Alzheimer disease is the most common neurodegenerative disorder and cause of senile dementia. It is characterized by an accelerated memory loss, and alterations of mood, reason, judgment and language. The main neuropathological hallmarks of the disorder are β-amyloid (βA) plaques and neurofibrillary Tau tangles. The triple transgenic 3xTgAD mouse model develops βA and Tau pathologies in a progressive manner which mimicks the pattern that takes place in the human brain with AD, and showing cognitive alterations characteristic of the disease. The present study intended to examine whether 3xTgAD mice of both sexes present cognitive, emotional and other behavioral alterations at the early age of 4 months, an age in which only some intraneuronal amyloid accumulation is found. Neonatal handling (H) is an early-life treatment known to produce profound and long-lasting behavioral and neurobiological effects in rodents, as well as improvements in cognitive functions. Therefore, we also aimed at evaluating the effects of H on the behavioral/cognitive profile of 4-month-old male and female 3xTgAD mice. The results indicate that, (1) 3xTgAD mice present spatial learning/memory deficits and emotional alterations already at the early age of 4 months, (2) there exists sexual dimorphism effects on several behavioral variables at this age, (3) neonatal handling exerts a preventive effect on some cognitive (spatial learning) and emotional alterations appearing in 3xTgAD mice already at early ages, and 4) H treatment appears to produce stronger positive effects in females than in males in several spatial learning measures and in the open field test.
Collapse
Affiliation(s)
- T Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| | - G Blázquez
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - L Giménez-Llort
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
38
|
Connors EJ, Shaik AN, Migliore MM, Kentner AC. Environmental enrichment mitigates the sex-specific effects of gestational inflammation on social engagement and the hypothalamic pituitary adrenal axis-feedback system. Brain Behav Immun 2014; 42:178-90. [PMID: 25011058 DOI: 10.1016/j.bbi.2014.06.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/27/2014] [Accepted: 06/27/2014] [Indexed: 10/25/2022] Open
Abstract
Modest environmental enrichment (EE) is well recognized to protect and rescue the brain from the consequences of a variety of insults. Although animal models of maternal immune activation (MIA) are associated with several neurodevelopmental impairments in both the behavioral and cognitive functioning of offspring, the impact of EE in protecting or reversing these effects has not been fully evaluated. In the present study, female Sprague-Dawley rats were randomized into EE (pair-housed in a large multi-level cage with toys, tubes and ramps) or animal care control (ACC; pair-housed in standard cages) conditions. Each pair was bred, following assignment to their housing condition, and administered 100μg/kg of lipopolysaccharide (LPS) on gestational day 11. After birth, and until the end of the study, offspring were maintained in their respective housing conditions. EE protected against both the social and hypothalamic pituitary adrenal axis consequences of MIA in juvenile male rats, but surprisingly not against the spatial discrimination deficits or accompanying decrease in glutamate levels within the hippocampus (as measured via LCMS-MS). Based on these preliminary results, the mechanisms that underlie the sex-specific consequences that follow MIA appear to be dependent on environmental context. Together, this work highlights the importance of environmental complexity in the prevention of neurodevelopmental deficits following MIA.
Collapse
Affiliation(s)
- E J Connors
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States
| | - A N Shaik
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| | - M M Migliore
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| | - A C Kentner
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States.
| |
Collapse
|
39
|
Melo AI. Role of sensory, social, and hormonal signals from the mother on the development of offspring. ADVANCES IN NEUROBIOLOGY 2014; 10:219-48. [PMID: 25287543 DOI: 10.1007/978-1-4939-1372-5_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
For mammals, sensory, social, and hormonal experience early in life is essential for the continuity of the infant's development. These experiences come from the mother through maternal care, and have enduring effects on the physiology and behavior of the adult organism. Disturbing the mother-offspring interaction by maternal deprivation (neglect) or exposure to adverse events as chronic stress, maltreatment, or sexual abuse has negative effects on the mental, psychological, physiological, and behavioral health. Indeed, these kinds of negative experiences can be the source of some neuropsychiatric diseases as depression, anxiety, impulsive aggression, and antisocial behavior. The purpose of this chapter is to review the most relevant evidence that supports the participation of cues from the mother and/or littermates during the postnatal preweaning period for the development of nervous system of the offspring. These findings come from the most frequently utilized experimental paradigms used in animal models, such as natural variations in maternal behavior, handling, partial maternal deprivation, and total maternal deprivation and artificial rearing. Through the use of these experimental procedures, it is possible to positively (handling paradigm), or negatively (maternal deprivation paradigms), affect the offspring's development. Finally, this chapter reviews the importance of the hormones that pups ingest through the maternal milk during early lactation on the development of several physiological systems, including the immune, endocrine systems, as well as on the adult behavior of the offspring.
Collapse
Affiliation(s)
- Angel I Melo
- Centro de Investigación en Reproducción Animal, CINVESTAV-Laboratorio Tlaxcala, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico,
| |
Collapse
|
40
|
Raineki C, Lucion AB, Weinberg J. Neonatal handling: an overview of the positive and negative effects. Dev Psychobiol 2014; 56:1613-25. [PMID: 25132525 DOI: 10.1002/dev.21241] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 06/24/2014] [Indexed: 02/06/2023]
Abstract
As one of the first rodent models designed to investigate the effects of early-life experiences, the neonatal handling paradigm has helped us better understand how subtle changes in the infant environment can powerfully drive neurodevelopment of the immature brain in typical or atypical trajectories. Here, we review data from more than 50 years demonstrating the compelling effects of neonatal handling on behavior, physiology, and neural function across the lifespan. Moreover, we present data that challenge the classical view of neonatal handling as an animal model that results only in positive/beneficial outcomes. Indeed, the overall goal of this review is to offer the suggestion that the effects of early-life experiences-including neonatal handling-are nuanced rather than unidirectional. Both beneficial and negative outcomes may occur, depending on the parameters of testing, sex of the subject, and neurobehavioral system analyzed.
Collapse
Affiliation(s)
- Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| | | | | |
Collapse
|
41
|
Roque S, Mesquita AR, Palha JA, Sousa N, Correia-Neves M. The behavioral and immunological impact of maternal separation: a matter of timing. Front Behav Neurosci 2014; 8:192. [PMID: 24904343 PMCID: PMC4033212 DOI: 10.3389/fnbeh.2014.00192] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 05/08/2014] [Indexed: 11/13/2022] Open
Abstract
Maternal separation (MS), an early life stressful event, has been demonstrated to trigger neuropsychiatric disorders later in life, in particular depression. Experiments using rodents subjected to MS protocols have been very informative for the establishment of this association. However, the mechanism by which MS leads to neuropsychiatric disorders is far from being understood. This is probably associated with the multifactorial nature of depression but also with the fact that different research MS protocols have been used (that vary on temporal windows and time of exposure to MS). In the present study, MS was induced in rats in two developmental periods: for 6 h per day for 14 days between postnatal days 2-15 (MS2-15) and 7-20 (MS7-20). These two periods were defined to differ essentially on the almost complete (MS2-15) or partial (MS7-20) overlap with the stress hypo-responsive period. Behavioral, immunological, and endocrine parameters, frequently associated with depressive-like behavior, were analyzed in adulthood. Irrespectively from the temporal window, both MS exposure periods led to increased sera corticosterone levels. However, only MS2-15 animals displayed depressive and anxious-like behaviors. Moreover, MS2-15 was also the only group presenting alterations in the immune system, displaying decreased percentage of CD8(+) T cells, increased spleen T cell CD4/CD8 ratio, and thymocytes with increased resistance to dexamethasone-induced cell death. A linear regression model performed to predict depressive-like behavior showed that both corticosterone levels and T cell CD4/CD8 ratio explained 37% of the variance observed in depressive-like behavior. Overall, these findings highlight the existence of "critical periods" for early life stressful events to exert programing effects on both central and peripheral systems, which are of relevance for distinct patterns of susceptibility to emotional disorders later in life.
Collapse
Affiliation(s)
- Susana Roque
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho , Braga , Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga , Portugal
| | - Ana Raquel Mesquita
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho , Braga , Portugal ; Neuropsychophysiology Laboratory, Center for Research in Psychology (CIPsi), School of Psychology, University of Minho , Braga , Portugal
| | - Joana A Palha
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho , Braga , Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga , Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho , Braga , Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga , Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho , Braga , Portugal ; ICVS/3B's - PT Government Associate Laboratory , Braga , Portugal
| |
Collapse
|
42
|
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
|
43
|
Nam H, Clinton SM, Jackson NL, Kerman IA. Learned helplessness and social avoidance in the Wistar-Kyoto rat. Front Behav Neurosci 2014; 8:109. [PMID: 24744709 PMCID: PMC3978372 DOI: 10.3389/fnbeh.2014.00109] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/14/2014] [Indexed: 12/31/2022] Open
Abstract
The Wistar-Kyoto (WKY) rat is an established depression model characterized by elevated anxiety- and depression-like behavior across a variety of tests. Here we further characterized specific behavioral and functional domains relevant to depression that are altered in WKY rats. Moreover, since early-life experience potently shapes emotional behavior, we also determined whether aspects of WKYs' phenotype were modifiable by early-life factors using neonatal handling or maternal separation. We first compared WKYs' behavior to that of Sprague–Dawley (SD), Wistar, and Spontaneously Hypertensive (SHR) rats in: the open field test, elevated plus maze, novelty-suppressed feeding test, a social interaction test, and the forced swim test (FST). WKYs exhibited high baseline immobility in the FST and were the only strain to show increased immobility on FST Day 2 vs. Day 1 (an indicator of learned helplessness). WKYs also showed greater social avoidance, along with enlarged adrenal glands and hearts relative to other strains. We next tested whether neonatal handling or early-life maternal separation stress influenced WKYs' behavior. Neither manipulation affected their anxiety- and depressive-like behaviors, likely due to a strong genetic underpinning of their phenotype. Our findings indicate that WKY rats are a useful model that captures specific functional domains relevant to clinical depression including: psychomotor retardation, behavioral inhibition, learned helplessness, social withdrawal, and physiological dysfunction. WKY rats appear to be resistant to early-life manipulations (i.e., neonatal handling) that are therapeutic in other strains, and may be a useful model for the development of personalized anti-depressant therapies for treatment resistant depression.
Collapse
Affiliation(s)
- Hyungwoo Nam
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA ; Cell, Molecular, and Developmental Biology, Graduate Biomedical Sciences Program, University of Alabama at Birmingham Birmingham, AL, USA
| | - Sarah M Clinton
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Nateka L Jackson
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Ilan A Kerman
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
44
|
da Silva Benetti C, Silveira PP, Wyse ATS, Scherer EBS, Ferreira AGK, Dalmaz C, Goldani MZ. Neonatal environmental intervention alters the vulnerability to the metabolic effects of chronic palatable diet exposure in adulthood. Nutr Neurosci 2014; 17:127-37. [PMID: 24621057 DOI: 10.1179/1476830513y.0000000077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Previous studies have demonstrated that early environmental interventions influence the consumption of palatable food and the abdominal fat deposition in female rats chronically exposed to a highly caloric diet in adulthood. In this study, we verified the metabolic effects of chronic exposure to a highly palatable diet, and determine the response to its withdrawal in adult neonatally handled and non-handled rats. Consumption of foods (standard lab chow and chocolate), body weight gain, abdominal fat deposition, plasma triglycerides, and leptin, as well as serum butyrylcholinesterase (BuChE), and cerebral acetylcholinesterase (AChE) activities were measured during chronic chocolate exposure and after deprivation of this palatable food in female rats exposed or not to neonatal handling (10 minutes/day, 10 first days of life). Handled rats increased rebound chocolate consumption in comparison to non-handled animals after 1 week of chocolate withdrawal; these animals also decreased body weight in the first 24 hours but this effect disappeared after 7 days of withdrawal. Chocolate increased abdominal fat in non-handled females, and this effect remained after 30 days of withdrawal; no differences in plasma leptin were seen after 7 days of withdrawal. Chocolate also increased serum BuChE activity in non-handled females, this effect was still evident after 7 days of withdrawal, but it disappeared after 30 days of withdrawal. Chocolate deprivation decreased cerebral AChE activity in both handled and non-handled animals. These findings suggest that neonatal handling modulates the preference for palatable food and induces a specific metabolic response that may be more adaptive in comparison to non-handled rats.
Collapse
|
45
|
Chokroborty-Hoque A, Alberry B, Singh SM. Exploring the complexity of intellectual disability in fetal alcohol spectrum disorders. Front Pediatr 2014; 2:90. [PMID: 25207264 PMCID: PMC4143882 DOI: 10.3389/fped.2014.00090] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 08/11/2014] [Indexed: 01/16/2023] Open
Abstract
Brain development in mammals is long lasting. It begins early during embryonic growth and is finalized in early adulthood. This progression represents a delicate choreography of molecular, cellular, and physiological processes initiated and directed by the fetal genotype in close interaction with environment. Not surprisingly, most aberrations in brain functioning including intellectual disability (ID) are attributed to either gene(s), or environment or the interaction of the two. The ensuing complexity has made the assessment of this choreography, ever challenging. A model to assess this complexity has used a mouse model (C57BL/6J or B6) that is subjected to prenatal alcohol exposure. The resulting pups show learning and memory deficits similar to patients with fetal alcohol spectrum disorder (FASD), which is associated with life-long changes in gene expression. Interestingly, this change in gene expression underlies epigenetic processes including DNA methylation and miRNAs. This paradigm is applicable to ethanol exposure at different developmental times (binge at trimesters 1, 2, and 3 as well as continuous preference drinking (70%) of 10% alcohol by B6 females during pregnancy). The exposure leads to life-long changes in neural epigenetic marks, gene expression, and a variety of defects in neurodevelopment and CNS function. We argue that this cascade may be reversed postnatally via drugs, chemicals, and environment including maternal care. Such conclusions are supported by two sets of results. First, antipsychotic drugs that are used to treat ID including psychosis function via changes in DNA methylation, a major epigenetic mark. Second, post-natal environment may improve (with enriched environments) or worsen (with negative and maternal separation stress) the cognitive ability of pups that were prenatally exposed to ethanol as well as their matched controls. In this review, we will discuss operational epigenetic mechanisms involved in the development of intellectual ability/disability in response to alcohol during prenatal or post-natal development. In doing so, we will explore the potential of epigenetic manipulation in the treatment of FASD and related disorders implicated in ID.
Collapse
Affiliation(s)
| | - Bonnie Alberry
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON , Canada
| | - Shiva M Singh
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON , Canada
| |
Collapse
|
46
|
George SA, Stout SA, Tan M, Knox D, Liberzon I. Early handling attenuates enhancement of glucocorticoid receptors in the prefrontal cortex in an animal model of post-traumatic stress disorder. BIOLOGY OF MOOD & ANXIETY DISORDERS 2013; 3:22. [PMID: 24289278 PMCID: PMC4175489 DOI: 10.1186/2045-5380-3-22] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/29/2013] [Indexed: 02/01/2023]
Abstract
BACKGROUND Changes in glucocorticoid receptors (GRs) have been implicated in the pathogenesis of stress related psychiatric disorders such as depression and post-traumatic stress disorder (PTSD). Abnormal adaptation of the stress-response system following traumatic stress can lead to an altered hypothalamic-pituitary-adrenal axis that may contribute to PTSD development. Indeed, elevated GR expression in the hippocampus and prefrontal cortex linked to PTSD-like characteristics have been reported in the validated animal model of PTSD, single-prolonged stress. These findings implicate increased levels of GRs in the development of post-traumatic psychopathology and suggest that exploration of GR-targeted interventions may have potential for PTSD prevention. Early handling during the neonatal phase alters GR expression and is proposed to confer resilience to stress. We therefore examined the effects of combined early handling and single prolonged stress treatments on GR expression. METHODS Timed pregnant dams gave birth to pups that were subjected to early handling (n = 11) or control (n = 13) procedures during the neonatal phase. At postnatal day 45 animals underwent single prolonged stress or a control procedure. Rats were euthanized one day later and GR levels were assayed using western blot electrophoresis. RESULTS Single prolonged stress exposure enhanced GR expression in the hippocampus and prefrontal cortex. Early handling treatment protected against single prolonged stress-induced enhancement of GR expression in the prefrontal cortex, but not in the hippocampus. CONCLUSIONS These data are a first step in highlighting the importance of targeting GR systems in prevention/resilience and may suggest that preventive strategies targeting GR upregulation might be particularly effective when prefrontal rather than hippocampal GRs are the target.
Collapse
Affiliation(s)
- Sophie A George
- Department of Psychiatry, University of Michigan, 4250 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Stephanie A Stout
- Department of Psychiatry, University of Michigan, 4250 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Melissa Tan
- Department of Psychiatry, University of Michigan, 4250 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Dayan Knox
- Department of Psychiatry, University of Michigan, 4250 Plymouth Road, Ann Arbor, MI 48109, USA
- Present address: Department of Psychology, University of Delaware, 108 Wolf Hall, Newark, DE 19716, USA
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, 4250 Plymouth Road, Ann Arbor, MI 48109, USA
- Ann Arbor Veterans Affairs Hospital, 2215 Fuller Road, Ann Arbor, MI 49105, USA
| |
Collapse
|
47
|
Brunton PJ. Effects of maternal exposure to social stress during pregnancy: consequences for mother and offspring. Reproduction 2013; 146:R175-89. [DOI: 10.1530/rep-13-0258] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A suboptimalin uteroenvironment, for example, as a result of maternal stress, can have detrimental effects on the pregnancy and long-term adverse ‘programming’ effects on the offspring. This article focuses on the effects of prenatal social stress on the mother, her pregnancy and the offspring, since these issues have ethological relevance in both animals and humans. The consequences of social stress exposure depend on when during pregnancy the stress occurs, and many of the effects on the offspring are sex specific. Social stress during early pregnancy tends to result in pregnancy loss, whereas stress exposure later in pregnancy, when the mother has already invested considerable resources in the foetuses, results in programmed offspring of low birth weight: a risk factor for various adulthood diseases. Neuroendocrine and behavioural responses to stress in the offspring are particularly sensitive to foetal programming by prenatal stress, indicated by enhanced hypothalamo-pituitary–adrenal (HPA) axis responses and increased anxiety behaviour, which result from permanent changes in the offspring's brain. The dysregulation of HPA axis function may also interfere with other systems, for example, the hypothalamic–pituitary–gonadal axis, as there is evidence for alterations in steroidogenesis, reproductive potential and impaired reproductive/social behaviours in prenatally stressed offspring. Prenatal social stress also programmes future maternal behaviour, highlighting the potential for negative phenotypes to be transmitted to future generations. The possible mechanisms through which maternal stress during pregnancy is transmitted to the foetuses and the foetal brain is programmed by prenatal stress and the potential to overwrite programming of the offspring are discussed.
Collapse
|
48
|
Clarke M, Cai G, Saleh S, Buller KM, Spencer SJ. Being suckled in a large litter mitigates the effects of early-life stress on hypothalamic-pituitary-adrenal axis function in the male rat. J Neuroendocrinol 2013; 25:792-802. [PMID: 23763285 DOI: 10.1111/jne.12056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/08/2013] [Accepted: 06/09/2013] [Indexed: 12/14/2022]
Abstract
The perinatal environment influences stress responses in the long-term, as does body composition. Male rats suckled in large litters, where they have reduced access to milk and attention from the dam, are less anxious and have attenuated hypothalamic-pituitary-adrenal (HPA) axis responses to stress compared to rats from control litters. In the present study, we investigated whether this early-life environment can also ameliorate anxiety and HPA axis function in rats prone to be stress-sensitive. We conducted these experiments in male rats from control litters (n = 12) or large litters (n = 20). Half were given 24 h of maternal separation on postnatal day 10 to induce HPA axis hyperactivity; the remainder staying undisturbed with their dam. When the rats reached adulthood, we examined behavioural indices of anxiety (elevated plus maze) and depression (Porsolt's forced swim test) under basal conditions and after 15 min of restraint stress. We also examined neuronal activation in the paraventricular nucleus of the hypothalamus (PVN) as an index of HPA axis function. Being suckled in a large litter led to a significantly attenuated PVN response to stress in adulthood. Maternal separation strongly exacerbated the stress-induced increase in PVN neuronal activation in control rats but did not affect the PVN response in large-litter rats. Immobility in the forced swim after restraint was also exacerbated in neonatally maternally separated control rats but not in those from large litters. Our findings show that being suckled in large litters mitigates the effects of early-life stress on HPA axis function and indices of depression in the rat.
Collapse
Affiliation(s)
- M Clarke
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
49
|
microRNAs and the regulation of neuronal plasticity under stress conditions. Neuroscience 2013; 241:188-205. [DOI: 10.1016/j.neuroscience.2013.02.065] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/26/2013] [Accepted: 02/26/2013] [Indexed: 12/21/2022]
|
50
|
Lynn SE, Kern MD, Phillips MM. Neonatal handling alters the development of the adrenocortical response to stress in a wild songbird (eastern bluebird, Sialia sialis). Gen Comp Endocrinol 2013; 186:157-63. [PMID: 23524000 DOI: 10.1016/j.ygcen.2013.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 03/02/2013] [Accepted: 03/04/2013] [Indexed: 02/01/2023]
Abstract
Neonatal handling of captive vertebrates can shape the development of their hypothalamo-pituitary-adrenal (HPA) axis and alter their ability to respond to stressful stimuli later in life. However, the long-term effects of such handling on this endocrine axis in free-living species are not well understood. We investigated the effects of age and neonatal handling on corticosterone secretion in response to restraint in eastern bluebird (Sialia sialis) chicks. We found that unhandled ("naïve") and handled ("experienced") chicks exhibited no corticosterone response to handling early in development. Thereafter, naïve individuals exhibited the progressive development of a corticosterone response with age, and by day 12 post-hatch, the response resembled that of adult bluebirds. Experienced nestlings, which were handled every other day from the day of hatch, showed a similar pattern of HPA development until day 12 post-hatch, when their corticosterone response was significantly reduced compared to that of naïve nestlings. In contrast, chicks that were handled only once, when 10days old, did not show a reduced corticosterone response at 12days old. Taken together, our data suggest that a certain threshold of accumulated neonatal handling episodes is necessary to depress corticosterone secretion, and/or that the cumulative effects of several handling episodes only manifest themselves once the HPA axis is fully developed. Our findings, in concert with studies on two other wild species, indicate that routine handling of nestlings in the field can alter their responses to stress in a species-specific manner, potentially leading to important fitness consequences.
Collapse
Affiliation(s)
- Sharon E Lynn
- The College of Wooster, Department of Biology, 931 College Mall, Wooster, OH 44691, USA.
| | | | | |
Collapse
|