1
|
Randell AM, Salia S, Fowler LF, Aung T, Puts DA, Swift-Gallant A. A meta-analysis of sex differences in neonatal rodent ultrasonic vocalizations and the implication for the preclinical maternal immune activation model. Biol Sex Differ 2025; 16:4. [PMID: 39863873 PMCID: PMC11762899 DOI: 10.1186/s13293-025-00685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
As the earliest measure of social communication in rodents, ultrasonic vocalizations (USVs) in response to maternal separation are critical in preclinical research on neurodevelopmental disorders (NDDs). While sex differences in both USV production and behavioral outcomes are reported, many studies overlook sex as a biological variable in preclinical NDD models. We aimed to evaluate sex differences in USV call parameters and determine if USVs are differently impacted based on sex in the preclinical maternal immune activation (MIA) model. Results indicate that sex differences in USVs vary with developmental stage and are more pronounced in MIA offspring. Specifically, developmental stage is a moderator of sex differences in USV call duration, with control females emitting longer calls than males in early development (up to postnatal day [PND] 8), but this pattern reverses after PND8. MIA leads to a reduction in call numbers for females compared to same-sex controls in early development, with a reversal post-PND8. MIA decreased call duration and increased total call duration in males, but unlike females, developmental stage did not influence these differences. In males, MIA effects varied by species, with decreased call numbers in rats but increased call numbers in mice. MIA timing (gestational day ≤ 12.5 vs. > 12.5) did not significantly affect results. Our findings highlight the importance of considering sex, developmental timing, and species in USVs research. We discuss how analyzing USV call types and incorporating sex as a biological variable can enhance our understanding of neonatal ultrasonic communication and its translational value in NDD research.
Collapse
Affiliation(s)
- Alison M Randell
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Lucas F Fowler
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
- Cognitive and Behavioural Ecology Program, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Toe Aung
- Department of Psychology and Counseling, Immaculata University, Immaculata, PA, USA
| | - David A Puts
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| |
Collapse
|
2
|
Hillerer KM, Gimsa U. Adult neurogenesis and the microbiota-gut-brain axis in farm animals: underestimated and understudied parameters for improving welfare in livestock farming. Front Neurosci 2024; 18:1493605. [PMID: 39664450 PMCID: PMC11631930 DOI: 10.3389/fnins.2024.1493605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Welfare in commercial livestock farming is becoming increasingly important in current agriculture research. Unfortunately, there is a lack of understanding about the neuronal mechanisms that underlie well-being on an individual level. Neuroplasticity in the hippocampus, the subventricular zone (SVZ), the olfactory bulb (OB) and the hypothalamus may be essential regulatory components in the context of farm animal behaviour and welfare that may be altered by providing environmental enrichment (EE). The importance of pre-and probiotics as a form of EE and the microbiota-gut-brain axis (MGBA) has come under the spotlight in the last 20 years, particularly in the contexts of research into stress and of stress resilience. However, it could also be an important regulatory system for animal welfare in livestock farming. This review aims to present a brief overview of the effects of EE on physiology and behaviour in farm animals and briefly discusses literature on behavioural flexibility, as well as inter-individual stress-coping styles and their relationship to animal welfare. Most importantly, we will summarise the literature on different forms of neural plasticity in farm animals, focusing on neurogenesis in various relevant brain regions. Furthermore, we will provide a brief outlook connecting these forms of neuroplasticity, stress, EE, the MGBA and welfare measures in modern livestock farming, concentrating on pigs.
Collapse
Affiliation(s)
- Katharina M. Hillerer
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Mecklenburg-Vorpommern, Germany
| | | |
Collapse
|
3
|
de Lima RMS, da Mata MJ, Santos JCPD, Costa L, Marques VHM, Bento LVDS, Lugon MDMV, Arcego DM, Barauna VG, Bittencourt AS, Bittencourt APSDV. Exploring the role of environmental enrichment and early life adversity on emotional development. Behav Brain Res 2024; 472:115147. [PMID: 39029628 DOI: 10.1016/j.bbr.2024.115147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/14/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Early life adversity has been linked with a higher probability of developing behavioral impairments and environmental manipulation is a strategy that may reduce the negative effects of exposure to adversity in early life. Here, we focused on exploring the influence of environmental enrichment (EE) as a protective factor in the context of early life adversity. We hypothesized that 24 hours of maternal deprivation (MD), in the second week of life, could induce anxiety-like behavior alterations and that exposure to EE could induce resilience to these behaviors due to alterations in the serotonergic system. Male Wistar rats were exposed to MD, on postnatal days 11 and 13, and to EE, after weaning. In adulthood, we performed a series of behavioral tests for fear, anxiety, and locomotor activity. We also measured the levels of serotonin in the amygdala and dorsal raphe nucleus. Our results revealed that MD does not impact fear behavior or the levels of serotonin, while EE decreases locomotor activity in a novel environment and enhances exploration in the predator odor test. EE also decreases serotonin in the amygdala and increases its turnover rate levels. Our findings provide insights into the critical timeframe during which stress exposure impacts the development and confirm that exposure to EE has an independent and protective effect for anxiety-like behaviors later in life.
Collapse
Affiliation(s)
- Randriely Merscher Sobreira de Lima
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil.
| | - Martielo Januario da Mata
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Josefa Cristina Pereira Dos Santos
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Ludhielle Costa
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Victor Hugo Moreira Marques
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Lucas Victor Dos Santos Bento
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Marcelo di Marcello Valladão Lugon
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Danusa Mar Arcego
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Valério Garrone Barauna
- Programa de Pós-Graduação em Ciências Fisiológicas, Centro de Ciências da Saúde, Departamento de Ciências Fisiológicas, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Athelson Stefanon Bittencourt
- Programa de Pós-Graduação em Bioquímica e Farmacologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Espírito Santo, Brazil; Departamento de Morfologia, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Ana Paula Santana de Vasconcellos Bittencourt
- Programa de Pós-Graduação em Ciências Fisiológicas, Centro de Ciências da Saúde, Departamento de Ciências Fisiológicas, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| |
Collapse
|
4
|
Mottarlini F, Rizzi B, Targa G, Buzzelli V, Di Trapano M, Rullo L, Candeletti S, Ciccocioppo R, Fattore L, Romualdi P, Fumagalli F, Trezza V, Caffino L. Communal nesting shapes the sex-dependent glutamatergic response to early life stress in the rat prefrontal cortex. Front Psychiatry 2024; 15:1406687. [PMID: 38835543 PMCID: PMC11148342 DOI: 10.3389/fpsyt.2024.1406687] [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: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction Early social environment, either positive or negative, shapes the adult brain. Communal nesting (CN), a naturalistic setting in which 2-3 females keep their pups in a single nest sharing care-giving behavior, provides high level of peer interaction for pups. Early social isolation (ESI) from dam and siblings represents, instead, an adverse condition providing no peer interaction. Methods We investigated whether CN (enrichment setting) might influence the response to ESI (impoverishment setting) in terms of social behavior and glutamate system in the medial prefrontal cortex (mPFC) of adult and adolescent male and female rats. Results Pinning (a rewarding component of social play behavior) was significantly more pronounced in males than in females exposed to the combination of CN and ESI. CN sensitized the glutamate synapse in the mPFC of ESI-exposed male, but not female, rats. Accordingly, we observed (i) a potentiation of the glutamatergic neurotransmission in the mPFC of both adolescent and adult males, as shown by the recruitment of NMDA receptor subunits together with increased expression/activation of PSD95, SynCAM 1, Synapsin I and αCaMKII; (ii) a de-recruiting of NMDA receptors from active synaptic zones of same-age females, together with reduced expression/activation of the above-mentioned proteins, which might reduce the glutamate transmission. Whether similar sex-dependent glutamate homeostasis modulation occurs in other brain areas remains to be elucidated. Discussion CN and ESI interact to shape social behavior and mPFC glutamate synapse homeostasis in an age- and sex-dependent fashion, suggesting that early-life social environment may play a crucial role in regulating the risk to develop psychopathology.
Collapse
Affiliation(s)
- Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
- Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Melania Di Trapano
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Liana Fattore
- Research National Council (CNR) Institute of Neuroscience-Cagliari, National Research Council, Cagliari, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, Istituto di Ricerca e Cura di Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Abstract
Early life experiences can have an enduring impact on the brain and behavior, with implications for stress reactivity, cognition, and social behavior. In particular, the neural systems that contribute to the expression of social behavior are altered by early life social environments. However, paradigms that have been used to alter the social environment during development have typically focused on exposure to stress, adversity, and deprivation of species-typical social stimulation. Here, we explore whether complex social environments can shape the development of complex social behavior. We describe lab-based paradigms for studying early life social complexity in rodents that are generally focused on enriching the social and sensory experiences of the neonatal and juvenile periods of development. The impact of these experiences on social behavior and neuroplasticity is highlighted. Finally, we discuss the degree to which our current approaches for studying social behavior outcomes give insight into "complex" social behavior and how social complexity can be better integrated into lab-based methodologies.
Collapse
Affiliation(s)
- James P Curley
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| | - Frances A Champagne
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
6
|
Bratzu J, Ciscato M, Pisanu A, Talani G, Frau R, Porcu P, Diana M, Fumagalli F, Romualdi P, Rullo L, Trezza V, Ciccocioppo R, Sanna F, Fattore L. Communal nesting differentially attenuates the impact of pre-weaning social isolation on behavior in male and female rats during adolescence and adulthood. Front Behav Neurosci 2023; 17:1257417. [PMID: 37915532 PMCID: PMC10616881 DOI: 10.3389/fnbeh.2023.1257417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Early social isolation (ESI) disrupts neurodevelopmental processes, potentially leading to long-lasting emotional and cognitive changes in adulthood. Communal nesting (CN), i.e., the sharing of parental responsibilities between multiple individuals in a nest, creates a socially enriching environment known to impact social and anxiety-related behaviors. Methods This study examines the effects of (i) the CN condition and of (ii) ESI during the 3rd week of life (i.e., pre-weaning ESI) on motor, cognitive, and emotional domains during adolescence and adulthood in male and female rats reared in the two different housing conditions, as well as (iii) the potential of CN to mitigate the impact of ESI on offspring. Results We found that in a spontaneous locomotor activity test, females exhibited higher activity levels compared to males. In female groups, adolescents reared in standard housing (SH) condition spent less time in the center of the arena, suggestive of increased anxiety levels, while the CN condition increased the time spent in the center during adolescence, but not adulthood, independently from ESI. The prepulse inhibition (PPI) test showed a reduced PPI in ESI adolescent animals of both sexes and in adult males (but not in adult females), with CN restoring PPI in males, but not in adolescent females. Further, in the marble burying test SH-ESI adolescent males exhibited higher marble burying behavior than all other groups, suggestive of obsessive-compulsive traits. CN completely reversed this stress-induced effect. Interestingly, ESI and CN did not have a significant impact on burying behavior in adult animals of both sexes. Discussion Overall, our findings (i) assess the effects of ESI on locomotion, sensorimotor gating, and compulsive-like behaviors, (ii) reveal distinct vulnerabilities of males and females within these domains, and (iii) show how early-life social enrichment may successfully counteract some of the behavioral alterations induced by early-life social stress in a sex-dependent manner. This study strengthens the notion that social experiences during early-life can shape emotional and cognitive outcomes in adulthood, and points to the importance of social enrichment interventions for mitigating the negative effects of early social stress on neurodevelopment.
Collapse
Affiliation(s)
- Jessica Bratzu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Maria Ciscato
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Augusta Pisanu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Giuseppe Talani
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Marco Diana
- G.Minardi’ Cognitive Neuroscience Laboratory, CPMB Science Department, University of Sassari, Sassari, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, University of Milan, Milan, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Viviana Trezza
- Department of Science, University “Roma Tre”, Rome, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Liana Fattore
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
7
|
Speck ML, Gomes ALA, Rojas CS, Willig JB, Herrmann AP, Pilger DA, Rates SMK. Environmental enrichment affects behavioral and pharmacological response to antidepressants in CF1 mice. Neurosci Lett 2023; 813:137432. [PMID: 37549865 DOI: 10.1016/j.neulet.2023.137432] [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: 04/18/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
It has been described that environmental enrichment (EE) exerts beneficial effects on cognitive and emotional performances, dendritic branching, synaptic density, neurogenesis and modulation of neurotrophic systems and neurotransmitters in rodents. However, the influence of EE on pharmacological and behavioral responses in animal models of psychiatric disorders has not been fully established. In this context, the aim of this study was to evaluate the influence of exposure to EE on mice behavior in the open field test (OFT) and forced swimming tests (FST), as well as the response to antidepressant drugs (fluoxetine 30 mg/kg and bupropion 30 mg/kg, p.o.). CF1 mice were exposed to an enriched housing condition at different developmental stages: from mating to postnatal day (PND) 55 (lifelong enrichment), from mating to PND21 (perinatal enrichment) and from PND21 to PND55 (post-weaning enrichment). At PND58 the male offspring were evaluated in the OFT and FST. BDNF gene expression in the hippocampus was determined through qPCR. Mice exposed to perinatal enrichment remained longer in the peripheral zone of the OFT and performed fewer grooming than mice housed under standard condition, and these effects were independent of drug treatment. Post-weaning and lifelong enrichment increased grooming behavior. Bupropion reduced grooming in all groups except in perinatal enriched. In turn, fluoxetine decreased grooming only in post-weaning enriched group. None of the enriched housing conditions altered the immobility time in the FST, which indicates that EE had no antidepressant-like effect. However, all enriched housing conditions abolished the anti-immobility effect of bupropion. None of the EE protocols affected BDNF hippocampal expression. The main conclusion is that mice behavior in the OFT is sensitive to alterations in the housing environment and depends on the developmental stage of exposure. Bupropion and fluoxetine yielded divergent responses depending on the housing condition, which suggests that EE modulates monoaminergic neurotransmission pathways.
Collapse
Affiliation(s)
- Marta Lorena Speck
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil
| | - Ana Luiza Azevedo Gomes
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil
| | - Camila Schafer Rojas
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil
| | - Julia Biz Willig
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil
| | - Ana Paula Herrmann
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics. Federal University of: Rio Grande do Sul. Rua Ramiro Barcelos, 2600, Porto Alegre, RS ZIP CODE 90035003, Brazil.
| | - Diogo André Pilger
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil.
| | - Stela Maris Kuze Rates
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul. Avenida Ipiranga, 2752, Porto Alegre, RS ZIP CODE 90610000, Brazil.
| |
Collapse
|
8
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
9
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
10
|
Liiver K, Imbeault S, Školnaja M, Kaart T, Kanarik M, Laugus K, De Wettinck J, Pulver A, Shimmo R, Harro J. Active vs passive novelty-related strategies: Sex differences in exploratory behaviour and monoaminergic systems. Behav Brain Res 2023; 441:114297. [PMID: 36641084 DOI: 10.1016/j.bbr.2023.114297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Sex differences are apparent in numerous behavioural characteristics. In order to compare and characterise male and female variability of exploratory behaviour, 365 male and 401 female rats were assessed in a task where a bimodal response distribution had previously been established in males. Female rats had significantly higher exploratory activity, and presented normal distribution of the behaviour, very differently from the bimodal distribution of males. No major effect of litter or oestrous cycle was detected. Several differences between male and female rats were found in monoamine metabolism measured ex vivo. Male rats had lower levels of dopamine (DA) in frontal cortex, and higher levels of 3,4-dihydroxyphenylacetic acid (DOPAC) in raphe area; higher levels of serotonin (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) in dorsal striatum but lower levels of 5-HT and 5-HIAA in locus coeruleus area, 5-HIAA levels were also lower in hippocampus as compared to females. Males had higher noradrenaline (NA) levels in hippocampus and lower normetanephrine (NMN) levels in striatum, in both brain regions male animals had lower NMN/NA ratio. No sex difference was found in accumbens. The only brain region with an interaction between sex and the expression of exploratory activity was raphe: Here 5-HT levels were lower, and DOPAC levels and DOPAC/DA and 5-HIAA/5-HT ratios higher in low exploring male but not female rats. Conclusively, female rats not only display higher levels of exploration but the population distribution of this behaviour is distinct; this may be related to differences in the monoaminergic systems between female and male animals.
Collapse
Affiliation(s)
- Kristi Liiver
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia
| | - Sophie Imbeault
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia
| | - Marianna Školnaja
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia; Laboratory Animal Centre, Tallinn University of Technology, Akadeemia Road 15, 12618 Tallinn, Estonia
| | - Tanel Kaart
- Institute of Veterinary Medicine and Animal Science, Estonian University of Life Sciences, 51006 Tartu, Estonia
| | - Margus Kanarik
- Division of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| | - Karita Laugus
- Division of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| | - Jade De Wettinck
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia
| | - Aleksander Pulver
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia
| | - Ruth Shimmo
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 25, 10120 Tallinn, Estonia; Division of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia; Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, 00014 Helsinki, Finland.
| |
Collapse
|
11
|
Corredor K, Duran J, Herrera-Isaza L, Forero S, Quintanilla J, Gomez A, Martínez GS, Cardenas FP. Behavioral effects of environmental enrichment on male and female wistar rats with early life stress experiences. Front Physiol 2022; 13:837661. [PMID: 36225294 PMCID: PMC9548697 DOI: 10.3389/fphys.2022.837661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to adverse childhood experiences or early life stress experiences (ELSs) increase the risk of non-adaptive behaviors and psychopathology in adulthood. Environmental enrichment (EE) has been proposed to minimize these effects. The vast number of methodological variations in animal studies underscores the lack of systematicity in the studies and the need for a detailed understanding of how enrichment interacts with other variables. Here we evaluate the effects of environmental enrichment in male and female Wistar rats exposed to adverse early life experiences (prenatal, postnatal, and combined) on emotional (elevated plus maze), social (social interaction chamber), memory (Morris water maze) and flexibility tasks. Our results—collected from PND 51 to 64—confirmed: 1) the positive effect of environmental enrichment (PND 28–49) on anxiety-like behaviors in animals submitted to ELSs. These effects depended on type of experience and type of enrichment: foraging enrichment reduced anxiety-like behaviors in animals with prenatal and postnatal stress but increased them in animals without ELSs. This effect was sex-dependent: females showed lower anxiety compared to males. Our data also indicated that females exposed to prenatal and postnatal stress had lower anxious responses than males in the same conditions; 2) no differences were found for social interactions; 3) concerning memory, there was a significant interaction between the three factors: A significant interaction for males with prenatal stress was observed for foraging enrichment, while physical enrichment was positive for males with postnatal stress; d) regarding cognitive flexibility, a positive effect of EE was found in animals exposed to adverse ELSs: animals with combined stress and exposed to physical enrichment showed a higher index of cognitive flexibility than those not exposed to enrichment. Yet, within animals with no EE, those exposed to combined stress showed lower flexibility than those exposed to both prenatal stress and no stress. On the other hand, animals with prenatal stress and exposed to foraging-type enrichment showed lower cognitive flexibility than those with no EE. The prenatal stress-inducing conditions used here 5) did not induced fetal or maternal problems and 6) did not induced changes in the volume of the dentate gyrus of the hippocampus.
Collapse
Affiliation(s)
- K. Corredor
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
- Centro de Investigación en Biomodelos, Bogotá, Colombia
| | - J.M. Duran
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - L. Herrera-Isaza
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - S. Forero
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - J.P. Quintanilla
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - A. Gomez
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | | | - F. P. Cardenas
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
- *Correspondence: F. P. Cardenas,
| |
Collapse
|
12
|
DeRosa H, Caradonna SG, Tran H, Marrocco J, Kentner AC. Milking It for All It's Worth: The Effects of Environmental Enrichment on Maternal Nurturance, Lactation Quality, and Offspring Social Behavior. eNeuro 2022; 9:ENEURO.0148-22.2022. [PMID: 35995560 PMCID: PMC9417599 DOI: 10.1523/eneuro.0148-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/10/2022] [Accepted: 07/15/2022] [Indexed: 12/17/2022] Open
Abstract
Breastfeeding confers robust benefits to offspring development in terms of growth, immunity, and neurophysiology. Similarly, improving environmental complexity, i.e., environmental enrichment (EE), contributes developmental advantages to both humans and laboratory animal models. However, the impact of environmental context on maternal care and milk quality has not been thoroughly evaluated, nor are the biological underpinnings of EE on offspring development understood. Here, Sprague Dawley rats were housed and bred in either EE or standard-housed (SD) conditions. EE dams gave birth to a larger number of pups, and litters were standardized and cross-fostered across groups on postnatal day (P)1. Maternal milk samples were then collected on P1 (transitional milk phase) and P10 (mature milk phase) for analysis. While EE dams spent less time nursing, postnatal enrichment exposure was associated with heavier offspring bodyweights. Milk from EE mothers had increased triglyceride levels, a greater microbiome diversity, and a significantly higher abundance of bacterial families related to bodyweight and energy metabolism. These differences reflected comparable transcriptomic changes at the genome-wide level. In addition to changes in lactational quality, we observed elevated levels of cannabinoid receptor 1 in the hypothalamus of EE dams, and sex-dependent and time-dependent effects of EE on offspring social behavior. Together, these results underscore the multidimensional impact of the combined neonatal and maternal environments on offspring development and maternal health. Moreover, they highlight potential deficiencies in the use of "gold standard" laboratory housing in the attempt to design translationally relevant animal models in biomedical research.
Collapse
Affiliation(s)
- Holly DeRosa
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| | | | - Hieu Tran
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| | - Jordan Marrocco
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
- Department of Biology, Touro University, New York, NY 10023
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| |
Collapse
|
13
|
Korgan AC, Foxx CL, Hashmi H, Sago SA, Stamper CE, Heinze JD, O'Leary E, King JL, Perrot TS, Lowry CA, Weaver ICG. Effects of paternal high-fat diet and maternal rearing environment on the gut microbiota and behavior. Sci Rep 2022; 12:10179. [PMID: 35715467 PMCID: PMC9205913 DOI: 10.1038/s41598-022-14095-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Exposing a male rat to an obesogenic high-fat diet (HFD) influences attractiveness to potential female mates, the subsequent interaction of female mates with infant offspring, and the development of stress-related behavioral and neural responses in offspring. To examine the stomach and fecal microbiome's potential roles, fecal samples from 44 offspring and stomach samples from offspring and their fathers were collected and bacterial community composition was studied by 16 small subunit ribosomal RNA (16S rRNA) gene sequencing. Paternal diet (control, high-fat), maternal housing conditions (standard or semi-naturalistic housing), and maternal care (quality of nursing and other maternal behaviors) affected the within-subjects alpha-diversity of the offspring stomach and fecal microbiomes. We provide evidence from beta-diversity analyses that paternal diet and maternal behavior induced community-wide shifts to the adult offspring gut microbiome. Additionally, we show that paternal HFD significantly altered the adult offspring Firmicutes to Bacteroidetes ratio, an indicator of obesogenic potential in the gut microbiome. Additional machine-learning analyses indicated that microbial species driving these differences converged on Bifidobacterium pseudolongum. These results suggest that differences in early-life care induced by paternal diet and maternal care significantly influence the microbiota composition of offspring through the microbiota-gut-brain axis, having implications for adult stress reactivity.
Collapse
Affiliation(s)
- Austin C Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Christine L Foxx
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Oak Ridge Institute for Science and Education Research Participation Program, Oak Ridge, TN, 37830, USA
- U.S. Department of Agriculture (USDA), National Animal Health Laboratory Network (NAHLN), Animal and Plant Health Inspection Service (APHIS), Ames, IA, 50010, USA
| | - Heraa Hashmi
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Saydie A Sago
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Christopher E Stamper
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Rocky Mountain MIRECC for Veteran Suicide Prevention, 1700 N Wheeling St, G-3-116M, Aurora, CO, 80045, USA
| | - Jared D Heinze
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Elizabeth O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jillian L King
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Department of Psychology and Neuroscience and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), The Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, 80045, USA
- Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO, 80045, USA
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Department of Psychiatry, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
14
|
Turner AD, Sullivan T, Drury K, Hall TA, Williams CN, Guilliams KP, Murphy S, Iqbal O’Meara AM. Cognitive Dysfunction After Analgesia and Sedation: Out of the Operating Room and Into the Pediatric Intensive Care Unit. Front Behav Neurosci 2021; 15:713668. [PMID: 34483858 PMCID: PMC8415404 DOI: 10.3389/fnbeh.2021.713668] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
In the midst of concerns for potential neurodevelopmental effects after surgical anesthesia, there is a growing awareness that children who require sedation during critical illness are susceptible to neurologic dysfunctions collectively termed pediatric post-intensive care syndrome, or PICS-p. In contrast to healthy children undergoing elective surgery, critically ill children are subject to inordinate neurologic stress or injury and need to be considered separately. Despite recognition of PICS-p, inconsistency in techniques and timing of post-discharge assessments continues to be a significant barrier to understanding the specific role of sedation in later cognitive dysfunction. Nonetheless, available pediatric studies that account for analgesia and sedation consistently identify sedative and opioid analgesic exposures as risk factors for both in-hospital delirium and post-discharge neurologic sequelae. Clinical observations are supported by animal models showing neuroinflammation, increased neuronal death, dysmyelination, and altered synaptic plasticity and neurotransmission. Additionally, intensive care sedation also contributes to sleep disruption, an important and overlooked variable during acute illness and post-discharge recovery. Because analgesia and sedation are potentially modifiable, understanding the underlying mechanisms could transform sedation strategies to improve outcomes. To move the needle on this, prospective clinical studies would benefit from cohesion with regard to datasets and core outcome assessments, including sleep quality. Analyses should also account for the wide range of diagnoses, heterogeneity of this population, and the dynamic nature of neurodevelopment in age cohorts. Much of the related preclinical evidence has been studied in comparatively brief anesthetic exposures in healthy animals during infancy and is not generalizable to critically ill children. Thus, complementary animal models that more accurately "reverse translate" critical illness paradigms and the effect of analgesia and sedation on neuropathology and functional outcomes are needed. This review explores the interactive role of sedatives and the neurologic vulnerability of critically ill children as it pertains to survivorship and functional outcomes, which is the next frontier in pediatric intensive care.
Collapse
Affiliation(s)
- Ashley D. Turner
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Travis Sullivan
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Kurt Drury
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Trevor A. Hall
- Department of Pediatrics, Division of Pediatric Psychology, Pediatric Critical Care and Neurotrauma Recovery Program, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Cydni N. Williams
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Kristin P. Guilliams
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
- Division of Pediatric Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Sarah Murphy
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - A. M. Iqbal O’Meara
- Department of Pediatrics, Child Health Research Institute, Children’s Hospital of Richmond at Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
15
|
Zhao X, Mohammed R, Tran H, Erickson M, Kentner AC. Poly (I:C)-induced maternal immune activation modifies ventral hippocampal regulation of stress reactivity: prevention by environmental enrichment. Brain Behav Immun 2021; 95:203-215. [PMID: 33766701 PMCID: PMC8187276 DOI: 10.1016/j.bbi.2021.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/14/2022] Open
Abstract
Environmental enrichment (EE) has been successfully implemented in human rehabilitation settings. However, the mechanisms underlying its success are not understood. Incorporating components of EE protocols into our animal models allows for the exploration of these mechanisms and their role in mitigation. Using a mouse model of maternal immune activation (MIA), the present study explored disruptions in social behavior and associated hypothalamic pituitary adrenal (HPA) axis functioning, and whether a supportive environment could prevent these effects. We show that prenatal immune activation of toll-like receptor 3, by the viral mimetic polyinosinic-polycytidylic acid (poly(I:C)), led to disrupted maternal care in that dams built poorer quality nests, an effect corrected by EE housing. Standard housed male and female MIA mice engaged in higher rates of repetitive rearing and had lower levels of social interaction, alongside sex-specific expression of several ventral hippocampal neural stress markers. Moreover, MIA males had delayed recovery of plasma corticosterone in response to a novel social encounter. Enrichment housing, likely mediated by improved maternal care, protected against these MIA-induced effects. We also evaluated c-Fos immunoreactivity associated with the novel social experience and found MIA to decrease neural activation in the dentate gyrus. Activation in the hypothalamus was blunted in EE housed animals, suggesting that the putative circuits modulating social behaviors may be different between standard and complex housing environments. These data demonstrate that augmentation of the environment supports parental care and offspring safety/security, which can offset effects of early health adversity by buffering HPA axis dysregulation. Our findings provide further evidence for the viability of EE interventions in maternal and pediatric settings.
Collapse
Affiliation(s)
| | | | | | | | - Amanda C. Kentner
- Corresponding author: Amanda Kentner, , Office #617-274-3360, Fax # 617-732-2959
| |
Collapse
|
16
|
The Contribution of Environmental Enrichment to Phenotypic Variation in Mice and Rats. eNeuro 2021; 8:ENEURO.0539-20.2021. [PMID: 33622702 PMCID: PMC7986535 DOI: 10.1523/eneuro.0539-20.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022] Open
Abstract
The reproducibility and translation of neuroscience research is assumed to be undermined by introducing environmental complexity and heterogeneity. Rearing laboratory animals with minimal (if any) environmental stimulation is thought to control for biological variability but may not adequately test the robustness of our animal models. Standard laboratory housing is associated with reduced demonstrations of species typical behaviors and changes in neurophysiology that may impact the translation of research results. Modest increases in environmental enrichment (EE) mitigate against insults used to induce animal models of disease, directly calling into question the translatability of our work. This may in part underlie the disconnect between preclinical and clinical research findings. Enhancing environmental stimulation for our model organisms promotes ethological natural behaviors but may simultaneously increase phenotypic trait variability. To test this assumption, we conducted a systematic review and evaluated coefficients of variation (CVs) between EE and standard housed mice and rats. Given findings of suboptimal reporting of animal laboratory housing conditions, we also developed a methodological reporting table for enrichment use in neuroscience research. Our data show that animals housed in EE were not more variable than those in standard housing. Therefore, environmental heterogeneity introduced into the laboratory, in the form of enrichment, does not compromise data integrity. Overall, human life is complicated, and by embracing such nuanced complexity into our laboratories, we may paradoxically improve on the rigor and reproducibility of our research.
Collapse
|
17
|
Sparling JE, Barbeau K, Boileau K, Konkle ATM. Environmental enrichment and its influence on rodent offspring and maternal behaviours, a scoping style review of indices of depression and anxiety. Pharmacol Biochem Behav 2020; 197:172997. [PMID: 32702399 DOI: 10.1016/j.pbb.2020.172997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022]
Abstract
Environmental enrichment is a widely used experimental manipulation that consistently shows measurable effects on rodent behaviour across the lifespan. This scoping review assesses and thematically summarizes the literature of the past decade concerning the effects of environmental enrichment applied during sensitive developmental periods in rodent mothers and offspring. Maternal behaviours as well as maternal and offspring anxiety- and depressive-like behaviours are considered. Relevant terms were searched across five databases (Embase, MEDLINE, PsycINFO, PubMed, Web of Science) and articles were screened with inclusion and exclusion criteria. The remaining articles were thematically analysed. Our results suggest that a greater number of articles reviewed the impacts of environmental enrichment on offspring anxiety-like behaviour (n = 23) rather than on depressive-like behaviour (n = 11) or maternal caregiving behaviour (n = 12). Maternal anxiety- (n = 4) or depressive-like (n = 2) behaviours are not often evaluated for in enrichment studies. The main behavioural tests of anxiety that were reviewed include the elevated plus-maze, the open field test, and the light-dark box whereas those for depression included the forced swim test and the sucrose preference test. Our results yielded mixed findings and significant variation in behavioural responses across all tests. In mothers, trends of increased maternal care behaviours and decreased maternal depressive-like behaviours in enriched mothers were appreciated. Enrichment during the gestational period was identified as pivotal to creating behavioural change in mother subjects. In enriched offspring rodents, a trend towards decreased anxiety-like behaviours was observed most often. Potential confounds inherent in enrichment paradigms and relevant theories of enrichment and their relation to rodent behavioural tests are discussed.
Collapse
Affiliation(s)
- Jessica E Sparling
- School of Psychology, University of Ottawa, Ottawa, Ontario, Canada; Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada.
| | - Kheana Barbeau
- School of Psychology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Kayla Boileau
- School of Psychology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Anne T M Konkle
- School of Psychology, University of Ottawa, Ottawa, Ontario, Canada; Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
18
|
Eck SR, Ardekani CS, Salvatore M, Luz S, Kim ED, Rogers CM, Hall A, Lee DE, Famularo ST, Bhatnagar S, Bangasser DA. The effects of early life adversity on growth, maturation, and steroid hormones in male and female rats. Eur J Neurosci 2020; 52:2664-2680. [PMID: 31660665 PMCID: PMC8027906 DOI: 10.1111/ejn.14609] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Early life adversity is a risk factor for psychiatric disorders, yet the mechanisms by which adversity increases this risk are still being delineated. Here, we used a limited bedding and nesting (LBN) manipulation in rats that models a low resource environment to examine effects on growth, developmental milestones, and endocrine endpoints. In LBN, dams and pups, from pups' postnatal days 2-9, are exposed to an environment where dams lack proper materials to build a nest. This manipulation is compared to control housing conditions, where rat dams have access to ample nesting materials and enrichment throughout pups' development. We found that the LBN condition altered maternal care, increasing pup-directed behaviors while reducing self-care. This, perhaps compensatory, increase in nursing and attention to pups did not mitigate against changes in metabolism, as LBN reduced weight gain in both sexes and this effect persisted into adulthood. Although adult stress hormone levels in both sexes and vaginal opening and estrous cycle length in females were not disrupted, there was other evidence of endocrine dysregulation. Compared to controls, LBN rats of both sexes had shortened anogenital distances, indicating reduced androgen exposure. LBN males also had higher plasma estradiol levels in adulthood. This combination of results suggests that LBN causes a demasculinizing effect in males that could contribute to lasting changes in the brain and behavior. Importantly, alterations in metabolic and endocrine systems due to early life adversity could be one mechanism by which stress early in life increases risk for later disease.
Collapse
Affiliation(s)
- Samantha R. Eck
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Cory S. Ardekani
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Madeleine Salvatore
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Sandra Luz
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eric D. Kim
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Charleanne M. Rogers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Arron Hall
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Demetrius E. Lee
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Sydney T. Famularo
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Debra A. Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
19
|
Baracz SJ, Everett NA, Robinson KJ, Campbell GR, Cornish JL. Maternal separation changes maternal care, anxiety-like behaviour and expression of paraventricular oxytocin and corticotrophin-releasing factor immunoreactivity in lactating rats. J Neuroendocrinol 2020; 32:e12861. [PMID: 32490585 DOI: 10.1111/jne.12861] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/31/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
The early postnatal period is a time of tremendous change for the dam and her offspring. During this time, environmental insults such as repeated stress exposure can have detrimental effects. In research that has focused on the effect of postnatal stress exposure on the dams, conflicting changes in maternal care and anxiety-like behaviour have been reported. Additionally, changes to hypothalamic neuropeptides that are crucially involved in the transition to motherhood and stress regulation, namely oxytocin and corticotrophin-releasing factor (CRF), have not been examined. Accordingly, the present study aimed to determine (i) whether repeated postpartum stress increases engagement in maternal care behaviours and anxiety-like behaviour and (ii) whether these behavioural changes correspond with changes to CRF- or oxytocin-immunoreactive (-IR) cells in the paraventricular nucleus (PVN) of the hypothalamus. A non-lactating group was also included to control for the effects of lactation on anxiety and the hypothalamic neuroendocrine system. Following the birth of their litters, Long-Evans dams were separated from their pups from postnatal day (PND) 1 to PND21 for either 15 minutes (maternal separation [MS]15) or 6 hours (MS360). Maternal behaviours were recorded for 30 minutes on select PNDs following the separation. On PND22, dams were exposed to the elevated plus maze, brains were collected, and immunofluorescence analysis of PVN oxytocin- and CRF-IR cells was conducted. Our findings demonstrate that prolonged maternal separation altered typical maternal behaviours and reduced anxiety relative to MS15 dams. At the cellular level, oxytocin-IR cells in the caudal PVN were reduced in MS360 dams to a level similar to that in non-lactating controls, and PVN CRF-IR cells were reduced relative to both MS15 and non-lactating controls. Taken together, these data reveal the behavioural and neuronal changes that occur in the mother dam following repeated postnatal stress exposure.
Collapse
Affiliation(s)
- Sarah J Baracz
- Department of Psychology, Macquarie University, North Ryde, NSW, Australia
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, Australia
| | - Nicholas A Everett
- Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | | | - Gemma R Campbell
- Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Jennifer L Cornish
- Department of Psychology, Macquarie University, North Ryde, NSW, Australia
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, Australia
| |
Collapse
|
20
|
Zhao X, Rondón-Ortiz AN, Lima EP, Puracchio M, Roderick RC, Kentner AC. Therapeutic efficacy of environmental enrichment on behavioral, endocrine, and synaptic alterations in an animal model of maternal immune activation. Brain Behav Immun Health 2020; 3. [PMID: 32368757 PMCID: PMC7197879 DOI: 10.1016/j.bbih.2020.100043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Maternal immune activation (MIA) has been identified as a significant risk factor for several neurodevelopmental disorders. We have previously demonstrated that postpubertal environmental enrichment (EE) rescues and promotes resiliency against MIA in male rats. Importantly, EE protocols have demonstrated clinical relevancy in human rehabilitation settings. Applying some of the elements of these EE protocols (e.g. social, physical, cognitive stimulation) to animal models of health and disease allows for the exploration of the mechanisms that underlie their success. Here, using a MIA model, we further investigate the rehabilitative potential of complex environments with a focus on female animals. Additionally, we expand upon some of our previous work by exploring genetic markers of synaptic plasticity and stress throughout several brain regions of both sexes. In the current study, standard housed female Sprague-Dawley rats were challenged with either the inflammatory endotoxin lipopolysaccharide (LPS; 100 μg/kg) or saline (equivolume) on gestational day 15. On postnatal day 50, male and female offspring were randomized into one of three conditions that differed in terms of cage size, number of cage mates (social stimulation) and enrichment materials. Spatial discrimination ability and social behavior were assessed six weeks later. Similar to our previously published work in males, our results revealed that a single LPS injection during mid gestation disrupted spatial discrimination ability in female rats. Postpubertal EE rescued this disruption. On the endocrine level, EE dampened elevations in plasma corticosterone that followed MIA, which may mediate EE's rehabilitative effects in female offspring. Within the prefrontal cortex, hippocampus, amygdala, and hypothalamus, MIA and EE altered the mRNA expression of several genes associated with resiliency and synaptic plasticity in both sexes. Overall, our findings provide further evidence that EE may serve as a therapeutic intervention for MIA-induced behavioral and cognitive deficits. Moreover, we identify some sexually dimorphic molecular mechanisms that may underlie these impairments and their rescue.
Collapse
Affiliation(s)
- Xin Zhao
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Alejandro N Rondón-Ortiz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Erika P Lima
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Madeline Puracchio
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Ryland C Roderick
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States 02115
| |
Collapse
|
21
|
Environmental influences on placental programming and offspring outcomes following maternal immune activation. Brain Behav Immun 2020; 83:44-55. [PMID: 31493445 PMCID: PMC6906258 DOI: 10.1016/j.bbi.2019.08.192] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/15/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Adverse experiences during pregnancy induce placental programming, affecting the fetus and its developmental trajectory. However, the influence of 'positive' maternal experiences on the placenta and fetus remain unclear. In animal models of early life stress, environmental enrichment (EE) has ameliorated and even prevented associated impairments in brain and behavior. Here, using a maternal immune activation (MIA) model in rats, we test whether EE attenuates maternal, placental and/or fetal responses to an inflammatory challenge, thereby offering a mechanism by which fetal programming may be prevented. Moreover, we evaluate life-long EE exposure on offspring development and examine a constellation of genes and epigenetic writers that may protect against MIA challenges. In our model, maternal plasma corticosterone and interleukin-1β were elevated 3 h after MIA, validating the maternal inflammatory response. Evidence for developmental programming was demonstrated by a simultaneous decrease in the placental enzymes Hsd11b2 and Hsd11b2/Hsd11b1, suggesting disturbances in glucocorticoid metabolism. Reductions of Hsd11b2 in response to challenge is thought to result in excess glucocorticoid exposure to the fetus and altered glucocorticoid receptor expression, increasing susceptibility to behavioral impairments later in life. The placental, but not maternal, glucocorticoid implications of MIA were attenuated by EE. There were also sustained changes in epigenetic writers in both placenta and fetal brain as a consequence of environmental experience and sex. Following MIA, both male and female juvenile animals were impaired in social discrimination ability. Life-long EE mitigated these impairments, in addition to the sex specific MIA associated disruptions in central Fkbp5 and Oprm1. These data provide the first evidence that EE protects placental functioning during stressor exposure, underscoring the importance of addressing maternal health and well-being throughout pregnancy. Future work must evaluate critical periods of EE use to determine if postnatal EE experience is necessary, or if prenatal exposure alone is sufficient to confer protection.
Collapse
|
22
|
Music exposure attenuates anxiety- and depression-like behaviors and increases hippocampal spine density in male rats. Behav Brain Res 2019; 372:112023. [DOI: 10.1016/j.bbr.2019.112023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/18/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
|
23
|
Gubert C, Hannan AJ. Environmental enrichment as an experience-dependent modulator of social plasticity and cognition. Brain Res 2019; 1717:1-14. [DOI: 10.1016/j.brainres.2019.03.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/11/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
|
24
|
Strzelewicz AR, Ordoñes Sanchez E, Rondón-Ortiz AN, Raneri A, Famularo ST, Bangasser DA, Kentner AC. Access to a high resource environment protects against accelerated maturation following early life stress: A translational animal model of high, medium and low security settings. Horm Behav 2019; 111:46-59. [PMID: 30708031 PMCID: PMC6527488 DOI: 10.1016/j.yhbeh.2019.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/18/2018] [Accepted: 01/10/2019] [Indexed: 12/21/2022]
Abstract
Early life exposure to a low security setting, characterized by a scarcity of resources and limited food access, increases the risk for psychiatric illness and metabolic dysfunction. We utilized a translational rat model to mimic a low security environment and determined how this manipulation affected offspring behavior, metabolism, and puberty. Because food insecurity in humans is associated with reduced access to healthy food options the "low security" rat manipulation combined a Western diet with exposure to a limited bedding and nesting manipulation (WD-LB). In this setting, dams were provided with limited nesting materials during the pups' early life (P2-P10). This manipulation was contrasted with standard rodent caging (SD) and environmental enrichment (EE), to model "medium security" and "high security" environments, respectively. To determine if transitioning from a low to high security environment improved outcomes, some juvenile WD-LB offspring were exposed to EE. Maternal care was impacted by these environments such that EE dams engaged in high quality care when on the nest, but spent less time on the nest than SD dams. Although WD-LB dams excessively chased their tails, they were very attentive to their pups, perhaps to compensate for limited resources. Offspring exposed to WD-LB only displayed subtle changes in behavior. However, WD-LB exposure resulted in significant metabolic dysfunction characterized by increased body weight, precocious puberty and alterations in the hypothalamic kisspeptin system. These negative effects of WD-LB on puberty and weight regulation were mitigated by EE exposure. Collectively, these studies suggest that both compensatory maternal care and juvenile enrichment can reduce the impact of a low security environment. Moreover, they highlight how utilizing diverse models of resource (in)stability can reveal mechanisms that confer vulnerability and resilience to early life stress.
Collapse
Affiliation(s)
- Arielle R Strzelewicz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | | | - Alejandro N Rondón-Ortiz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | - Anthony Raneri
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Sydney T Famularo
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Debra A Bangasser
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States.
| |
Collapse
|
25
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
26
|
Kokras N, Sotiropoulos I, Besinis D, Tzouveka EL, Almeida OFX, Sousa N, Dalla C. Neuroplasticity-related correlates of environmental enrichment combined with physical activity differ between the sexes. Eur Neuropsychopharmacol 2019; 29:1-15. [PMID: 30497839 DOI: 10.1016/j.euroneuro.2018.11.1107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 11/01/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
Abstract
Environmental enrichment (EE), comprising positive physical (exercise) and cognitive stimuli, influences neuronal structure and usually improves brain function. The promise of EE as a preventative strategy against neuropsychiatric disease is especially high during early postnatal development when the brain is still amenable to reorganization. Despite the fact that male and female brains differ in terms of connectivity and function that may reflect early life experiences, knowledge of the neural substrates and mechanisms by which such changes arise remains limited. This study compared the impact of EE combined with physical activity on neuroplasticity and its functional consequences in adult male and female rats; EE was provided during the first 3 months of life and our analysis focused on the hippocampus, an area implicated in cognitive behavior as well as the neuroendocrine response to stress. Both male and female rats reared in EE displayed better object recognition memory than their control counterparts. Interestingly, sex differences were revealed in the effects of EE on time spent exploring the objects during this test. Independently of sex, EE increased hippocampal turnover rates of dopamine and serotonin and reduced expression of 5-HT1A receptors; in addition, EE upregulated expression of synaptophysin, a presynaptic protein, in the hippocampus. As compared to their respective controls, EE-exposed males exhibited parallel increases in phosphorylated Tau and the GluN2B receptor, whereas females responded to EE with reduced hippocampal levels of glutamate and GluN2B. Together, these observations provide further evidence on the differential effects of EE on markers of hippocampal neuroplasticity in males and females.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece; First Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - I Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - D Besinis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - E L Tzouveka
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | | | - N Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece.
| |
Collapse
|
27
|
Kentner AC, Bilbo SD, Brown AS, Hsiao EY, McAllister AK, Meyer U, Pearce BD, Pletnikov MV, Yolken RH, Bauman MD. Maternal immune activation: reporting guidelines to improve the rigor, reproducibility, and transparency of the model. Neuropsychopharmacology 2019; 44:245-258. [PMID: 30188509 PMCID: PMC6300528 DOI: 10.1038/s41386-018-0185-7] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/04/2018] [Accepted: 08/02/2018] [Indexed: 01/16/2023]
Abstract
The 2017 American College of Neuropychopharmacology (ACNP) conference hosted a Study Group on 4 December 2017, Establishing best practice guidelines to improve the rigor, reproducibility, and transparency of the maternal immune activation (MIA) animal model of neurodevelopmental abnormalities. The goals of this session were to (a) evaluate the current literature and establish a consensus on best practices to be implemented in MIA studies, (b) identify remaining research gaps warranting additional data collection and lend to the development of evidence-based best practice design, and (c) inform the MIA research community of these findings. During this session, there was a detailed discussion on the importance of validating immunogen doses and standardizing the general design (e.g., species, immunogenic compound used, housing) of our MIA models both within and across laboratories. The consensus of the study group was that data does not currently exist to support specific evidence-based model selection or methodological recommendations due to lack of consistency in reporting, and that this issue extends to other inflammatory models of neurodevelopmental abnormalities. This launched a call to establish a reporting checklist focusing on validation, implementation, and transparency modeled on the ARRIVE Guidelines and CONSORT (scientific reporting guidelines for animal and clinical research, respectively). Here we provide a summary of the discussions in addition to a suggested checklist of reporting guidelines needed to improve the rigor and reproducibility of this valuable translational model, which can be adapted and applied to other animal models as well.
Collapse
Affiliation(s)
- Amanda C. Kentner
- 0000 0001 0021 3995grid.416498.6School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA USA
| | - Staci D. Bilbo
- 000000041936754Xgrid.38142.3cDepartment of Pediatrics, Harvard Medical School, Boston, MA USA ,0000 0004 0386 9924grid.32224.35Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA USA
| | - Alan S. Brown
- 0000000419368729grid.21729.3fDepartment of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, NY USA
| | - Elaine Y. Hsiao
- 0000 0000 9632 6718grid.19006.3eDepartment of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - A. Kimberley McAllister
- 0000 0004 1936 9684grid.27860.3bCenter for Neuroscience, University of California Davis, Davis, CA USA
| | - Urs Meyer
- 0000 0004 1937 0650grid.7400.3Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse, Zurich, Switzerland ,0000 0004 1937 0650grid.7400.3Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Brad D. Pearce
- 0000 0001 0941 6502grid.189967.8Department of Epidemiology, Rollins School of Public Health, and Graduate Division of Biological and Biomedical Sciences, Neuroscience Program, Emory University, Atlanta, GA USA
| | - Mikhail V. Pletnikov
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry and Behavioral Sciences, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Robert H. Yolken
- 0000 0001 2171 9311grid.21107.35Department of Pediatrics, Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Melissa D. Bauman
- 0000 0004 1936 9684grid.27860.3bThe UC Davis MIND Institute, Department of Psychiatry and Behavioral Sciences, California National Primate Research Center, University of California, Davis, USA
| |
Collapse
|
28
|
Cutuli D, Berretta E, Laricchiuta D, Caporali P, Gelfo F, Petrosini L. Pre-reproductive Parental Enriching Experiences Influence Progeny's Developmental Trajectories. Front Behav Neurosci 2018; 12:254. [PMID: 30483072 PMCID: PMC6240645 DOI: 10.3389/fnbeh.2018.00254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023] Open
Abstract
While the positive effects of environmental enrichment (EE) applied after weaning, in adulthood, during aging, or even in the presence of brain damage have been widely described, the transgenerational effects of pre-reproductive EE have been less examined. And yet, this issue is remarkable given that parental environmental experience may imprint offspring’s phenotype over generations through many epigenetic processes. Interactions between individual and environment take place lifelong even before conception. In fact, the environment pre-reproductively experienced by the mother and/or the father exerts a substantial impact on neural development and motor and cognitive performances of the offspring, even if not directly exposed to social, cognitive, physical and/or motor enrichment. Furthermore, pre-reproductive parental enrichment exerts a transgenerational impact on coping response to stress as well as on the social behavior of the offspring. Among the effects of pre-reproductive parental EE, a potentiation of the maternal care and a decrease in global methylation levels in the frontal cortex and hippocampus of the progeny have been described. Finally, pre-reproductive EE modifies different pathways of neuromodulation in the brain of the offspring (involving brain-derived neurotrophic factor, oxytocin and glucocorticoid receptors). The present review highlights the importance of pre-reproductive parental enrichment in altering the performances not only of animals directly experiencing it, but also of their progeny, thus opening the way to new hypotheses on the inheritance mechanisms of behavioral traits.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Erica Berretta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Daniela Laricchiuta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Paola Caporali
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Francesca Gelfo
- Fondazione Santa Lucia, Rome, Italy.,Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | - Laura Petrosini
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
29
|
Sparling JE, Baker SL, Bielajew C. Effects of combined pre- and post-natal enrichment on anxiety-like, social, and cognitive behaviours in juvenile and adult rat offspring. Behav Brain Res 2018; 353:40-50. [DOI: 10.1016/j.bbr.2018.06.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/22/2018] [Accepted: 06/28/2018] [Indexed: 01/14/2023]
|
30
|
Lopes DA, Souza TM, de Andrade JS, Silva MF, Antunes HK, Sueur-Maluf LL, Céspedes IC, Viana MB. Environmental enrichment decreases avoidance responses in the elevated T-maze and delta FosB immunoreactivity in anxiety-related brain regions. Behav Brain Res 2018; 344:65-72. [DOI: 10.1016/j.bbr.2018.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 01/16/2023]
|
31
|
Korgan AC, O'Leary E, King JL, Weaver ICG, Perrot TS. Effects of paternal high-fat diet and rearing environment on maternal investment and development of defensive responses in the offspring. Psychoneuroendocrinology 2018. [PMID: 29518693 DOI: 10.1016/j.psyneuen.2018.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Paternal preconception risk factors (e.g. stress, diet, drug use) correlate with metabolic dysfunction in offspring, which is often comorbid with depressive and anxiety-like phenotypes. Detection of these risk factors or deleterious phenotypes informs a female about prevailing ecological demands, in addition to potential adverse environment-induced phenotypes that may be disseminated to her offspring. We examined whether a F0 male rat's prior exposure to an obesogenic high-fat diet (HFD) influences a female's attraction towards a male, subsequent mother-infant interactions and the development of defensive (emotional) responses in the F1 offspring. Females displayed less interest in the HFD exposed F0 males relative to control diet-exposed F0 males. Dams that reared F1 offspring in larger, semi-naturalistic housing provided more licking and grooming and active arched-back-nursing behavior. However, some of these effects interacted with paternal experience. F0 HFD and maternal rearing environment revealed sex-dependent, between group differences in F1 offspring wean weight, juvenile social interactions and anxiety-like behavior in adolescence. Our results show for the first time in mammals that male exposure to HFD may contribute to stable behavioral variation among females in courtship, maternal care, even when the females are not directly exposed to a HFD, and anxiety-like behavior in F1 offspring. Furthermore, when offspring were exposed to a predatory threat, hypothalamic Crf gene regulation was influenced by early housing. These results, together with our previous findings, suggest that paternal experience and maternal rearing conditions can influence maternal behavior and development of defensive responses of offspring.
Collapse
Affiliation(s)
- Austin C Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Elizabeth O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Jillian L King
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada.
| |
Collapse
|
32
|
Cutuli D, Berretta E, Caporali P, Sampedro-Piquero P, De Bartolo P, Laricchiuta D, Gelfo F, Pesoli M, Foti F, Farioli Vecchioli S, Petrosini L. Effects of pre-reproductive maternal enrichment on maternal care, offspring's play behavior and oxytocinergic neurons. Neuropharmacology 2018; 145:99-113. [PMID: 29462694 DOI: 10.1016/j.neuropharm.2018.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 01/10/2023]
Abstract
Potentiating social, cognitive, and sensorimotor stimulations the Environmental Enrichment (EE) increases levels of novelty and complexity experienced by individuals. Growing evidence demonstrates that parental EE experience, even occurring in the pre-reproductive phase, affects behavioral and neural developmental trajectories of the offspring. To discover how the accumulation of early maternal complex experiences may inform and shape the social behavior of the following generation, we examined the effects of pre-reproductive enrichment of dams (post-natal days 21-72) on the play performances of their male and female adolescent offspring. Furthermore, we examined the effects of pre-reproductive enrichment on maternal behavior (during post-partum days 1-10) and male intruder aggression (on post-partum day 11). Since oxytocin modulates maternal care, social bonding, and agonistic behavior, the number of oxytocinergic neurons of the paraventricular (PVN) and supraoptic (SON) nuclei was examined in both dams and offspring. Results revealed that enriched females exhibited higher levels of pup-oriented behaviors, especially Crouching, and initiated pup-retrieval more quickly than standard females after the maternal aggression test. Such behavioral peculiarities were accompanied by increased levels of oxytocinergic neurons in PVN and SON. Moreover, pre-reproductive maternal EE cross-generationally influenced the offspring according to sex. Indeed, male pups born to enriched females exhibited a reduced play fighting associated with a higher number of oxytocinergic neurons in SON in comparison to male pups born to standard-housed females. In conclusion, pre-reproductive EE to the mothers affects their maternal care and has a cross-generational impact on the social behavior of their offspring that do not directly experiences EE. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; Fondazione Santa Lucia, Rome, Italy.
| | - Erica Berretta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; Fondazione Santa Lucia, Rome, Italy
| | - Paola Caporali
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Patricia Sampedro-Piquero
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento. Facultad de Psicología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Spain
| | - Paola De Bartolo
- Fondazione Santa Lucia, Rome, Italy; Department of TeCoS, Guglielmo Marconi University, Rome, Italy
| | - Daniela Laricchiuta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; Fondazione Santa Lucia, Rome, Italy
| | - Francesca Gelfo
- Fondazione Santa Lucia, Rome, Italy; Department of TeCoS, Guglielmo Marconi University, Rome, Italy
| | - Matteo Pesoli
- Fondazione Santa Lucia, Rome, Italy; Department of Motor Science and Wellness, University Parthenope, Naples, Italy
| | - Francesca Foti
- Fondazione Santa Lucia, Rome, Italy; Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | | | - Laura Petrosini
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
33
|
Complex Environmental Rearing Enhances Social Salience and Affects Hippocampal Corticotropin Releasing Hormone Receptor Expression in a Sex-Specific Manner. Neuroscience 2017; 369:399-411. [PMID: 29183827 DOI: 10.1016/j.neuroscience.2017.11.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/27/2017] [Accepted: 11/17/2017] [Indexed: 12/26/2022]
Abstract
Methods for understanding the neurocircuitry of ethologically relevant behaviors have advanced substantially; however renovations to standard animal laboratory housing, in the form of enhanced enrichment, have lagged behind. This is despite evidence that environmental enrichment (EE) reduces stress, stereotypy, and promotes healthy species typical behaviors. While many scientists express interest for increased EE as a standard for animal caging systems, there are concerns that its effects on brain, behavior, and cognition are not well characterized. In the present study, male and female Sprague-Dawley rats were housed for six weeks in either EE, Colony Nesting (CN), or Standard Housing (SD) conditions. We show that adolescent exposure to environmental complexity changed the dynamics of social interactions, sensory processing, and underlying basal stress neurocircuitry, in a sex- and enrichment-type-dependent manner. Specifically, EE and CN increased prosocial engagement and the social saliency of male and female rats while the profile of hippocampal Crhr2 expression was affected only in EE males. Hippocampal Crh was associated with anxiety-like behavior in SD males - this did not extend to EE or CN groups, nor to females. Observations such as these are an important consideration for the validity of translational research investigating the neurocircuitry of stress resiliency, and for understanding the mechanisms of psychiatric disorders. Future work must focus on characterizing how individual environmental enhancements (e.g. novelty, social enrichment, physical activity) shape phenotypic differences, how they vary as a function of species, strain and sex, and (if warranted) how to meaningfully implement this knowledge into biomedical research designs.
Collapse
|
34
|
Green A, Esser MJ, Perrot TS. Developmental expression of anxiety and depressive behaviours after prenatal predator exposure and early life homecage enhancement. Behav Brain Res 2017; 346:122-136. [PMID: 29183765 DOI: 10.1016/j.bbr.2017.11.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 01/25/2023]
Abstract
Stressful events during gestation can have sex-specific effects on brain and behaviour, and may contribute to some of the differences observed in adult stress responding and psychopathology. We investigated the impact of a novel repeated prenatal psychological stress (prenatal predator exposure - PPS) during the last week of gestation in rats on offspring behaviours related to social interaction (play behaviour), open field test (OFT), forced swim test (FST) and sucrose preference test (SP) during the juvenile period and in adulthood. We further examined the role of postnatal environmental, using an enhanced housing condition (EHC), to prevent/rescue any changes. Some effects on anxiety, anhedonia, and stress-related coping behaviours (e.g., OFT, SP and OFT) did not emerge until adulthood. PPS increased OFT anxiety behaviours in adult males, and some OFT and SP behaviours in adult females. Contrary to this, EHC had few independent effects; most were apparent only when combined with PPS. In keeping with age-group differences, juvenile behaviours did not necessarily predict the same adult behaviours although juvenile OFT rearing and freezing, and juvenile FST immobility did predict adult FST immobility and sucrose preference, suggesting that some aspects of depressive behaviours may emerge early and predict adult vulnerability or coping behaviours. Together, these results suggest an important, though complex, role for early life psychological stressors and early life behaviours in creating an adult vulnerability to anxiety or depressive disorders and that environmental factors further modulate the effects of the prenatal stressors.
Collapse
Affiliation(s)
- Amanda Green
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Michael J Esser
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
35
|
Yan S, Kentner AC. Mechanical allodynia corresponds to Oprm1 downregulation within the descending pain network of male and female rats exposed to neonatal immune challenge. Brain Behav Immun 2017; 63:148-159. [PMID: 27742580 DOI: 10.1016/j.bbi.2016.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/31/2022] Open
Abstract
Exposure to painful procedures and/or stressors during the early neonatal period can reprogram the underlying neurocircuitry involved in nociception and neuropathic pain perception. The reprogramming of these systems can result in an enduring elevation in sensitivity towards mechanical and thermal stimuli. Recent evidence suggests that exposure to mild inflammatory mediators during the neonatal period can induce similar pain responses in both adolescent and adult rats. Therefore, we sought to profile changes in the expression of several genes across brain areas involved in the active modulation of nociception and neuropathic pain using a well-recognized model of neonatal inflammation. In the present study male and female Sprague-Dawley rats were administered either the inflammatory endotoxin lipopolysaccharide (LPS; 0.05mg/kg, i.p.) or saline (equivolume) on postnatal days (PND) 3 and 5. During adolescence, hind paw mechanical withdrawal thresholds were evaluated using an electronic von Frey anesthesiometer. Animals challenged neonatally with LPS (nLPS) had increased pain sensitivity on this measure which was associated with decreased Oprm1 expression in the prefrontal cortex (PFC) and periaqueductal gray (PAG) of both male and female rats. Although a 'second hit' with LPS in adolescence (aLPS) did not confer protection or reveal additional vulnerabilities, aLPS given to animals treated neonatally with saline was associated with increased pain sensitivity, but only in females. Interestingly, adolescent inflammatory challenge decreased Hcrt2 mRNA in the PAG and elevated Trpv1 in the PAG and PFC of both sexes. There was no effect of inflammatory treatment on either anxiety or depressive-like behavior suggesting that affective functioning did not account for differences in mechanical pain sensitivity. Finally, a preliminary investigation demonstrated that administration of a broad spectrum antibiotic cocktail attenuated the mechanical sensitivity that followed nLPS. Together, these data extend upon evidence that inflammation imparts long term changes in quality of life and pain responses via interference within the descending pain network. Moreover, they highlight a potential window of opportunity to target the microbiota-gut-brain axis and reverse pain processing disturbances following perinatal inflammation.
Collapse
Affiliation(s)
- Siyang Yan
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States.
| |
Collapse
|
36
|
Durán-Carabali LE, Arcego DM, Odorcyk FK, Reichert L, Cordeiro JL, Sanches EF, Freitas LD, Dalmaz C, Pagnussat A, Netto CA. Prenatal and Early Postnatal Environmental Enrichment Reduce Acute Cell Death and Prevent Neurodevelopment and Memory Impairments in Rats Submitted to Neonatal Hypoxia Ischemia. Mol Neurobiol 2017; 55:3627-3641. [DOI: 10.1007/s12035-017-0604-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
|
37
|
Korgan AC, O’Leary E, Bauer J, Fortier A, Weaver ICG, Perrot TS. Effects of Paternal Predation Risk and Rearing Environment on Maternal Investment and Development of Defensive Responses in the Offspring. eNeuro 2016; 3:ENEURO.0231-16.2016. [PMID: 27896313 PMCID: PMC5112542 DOI: 10.1523/eneuro.0231-16.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/19/2016] [Accepted: 11/01/2016] [Indexed: 01/05/2023] Open
Abstract
Detecting past experiences with predators of a potential mate informs a female about prevailing ecological threats, in addition to stress-induced phenotypes that may be disseminated to offspring. We examined whether prior exposure of a male rat to a predator (cat) odor influences the attraction of a female toward a male, subsequent mother-infant interactions and the development of defensive (emotional) responses in the offspring. Females displayed less interest in males that had experienced predator odor. Mothers that reared young in larger, seminaturalistic housing provided more licking and grooming and active arched back-nursing behavior toward their offspring compared with dams housed in standard housing, although some effects interacted with paternal experience. Paternal predation risk and maternal rearing environment revealed sex-dependent differences in offspring wean weight, juvenile social interactions, and anxiety-like behavior in adolescence. Additionally, paternal predator experience and maternal housing independently affected variations in crf gene promoter acetylation and crf gene expression in response to an acute stressor in offspring. Our results show for the first time in mammals that variation among males in their predator encounters may contribute to stable behavioral variation among females in preference for mates and maternal care, even when the females are not directly exposed to predator threat. Furthermore, when offspring were exposed to the same threat experienced by the father, hypothalamic crf gene regulation was influenced by paternal olfactory experience and early housing. These results, together with our previous findings, suggest that paternal stress exposure and maternal rearing conditions can influence maternal behavior and the development of defensive responses in offspring.
Collapse
Affiliation(s)
- Austin C. Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Elizabeth O’Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Jessica Bauer
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Aidan Fortier
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Ian C. G. Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
- Department of Psychiatry, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
- Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Tara S. Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
- Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| |
Collapse
|
38
|
Kentner AC, Khoury A, Lima Queiroz E, MacRae M. Environmental enrichment rescues the effects of early life inflammation on markers of synaptic transmission and plasticity. Brain Behav Immun 2016; 57:151-160. [PMID: 27002704 DOI: 10.1016/j.bbi.2016.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/04/2016] [Accepted: 03/17/2016] [Indexed: 12/13/2022] Open
Abstract
Environmental enrichment (EE) has been successful at rescuing the brain from a variety of early-life psychogenic stressors. However, its ability to reverse the behavioral and neural alterations induced by a prenatal maternal infection model of schizophrenia is less clear. Moreover, the specific interactions between the components (i.e. social enhancement, novelty, physical activity) of EE that lead to its success as a supportive intervention have not been adequately identified. In the current study, standard housed female Sprague-Dawley rats were administered either the inflammatory endotoxin lipopolysaccharide (LPS; 100μg/kg) or pyrogen-free saline (equivolume) on gestational day 15. On postnatal day 50, offspring were randomized into one of three conditions: EE (group housed in a large multi-level cage with novel toys, tubes and ramps), Colony Nesting (CN; socially-housed in a larger style cage), or Standard Care (SC; pair-housed in standard cages). Six weeks later we scored social engagement and performance in the object-in-place task. Afterwards hippocampus and prefrontal cortex (n=7-9) were collected and evaluated for excitatory amino acid transporter (EAAT) 1-3, brain-derived neurotrophic factor (BDNF), and neurotrophic tyrosine kinase, receptor type 2 (TrkB) gene expression (normalized to GAPDH) using qPCR methods. Overall, we show that gestational inflammation downregulates genes critical to synaptic transmission and plasticity, which may underlie the pathogenesis of neurodevelopmental disorders such as schizophrenia and autism. Additionally, we observed disruptions in both social engagement and spatial discrimination. Importantly, behavioral and neurophysiological effects were rescued in an experience dependent manner. Given the evidence that schizophrenia and autism may be associated with infection during pregnancy, these data have compelling implications for the prevention and reversibility of the consequences that follow immune activation in early in life.
Collapse
Affiliation(s)
- Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly the Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States.
| | - Antoine Khoury
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| | | | - Molly MacRae
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly the Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States
| |
Collapse
|
39
|
Zuena AR, Zinni M, Giuli C, Cinque C, Alemà GS, Giuliani A, Catalani A, Casolini P, Cozzolino R. Maternal exposure to environmental enrichment before and during gestation influences behaviour of rat offspring in a sex-specific manner. Physiol Behav 2016; 163:274-287. [DOI: 10.1016/j.physbeh.2016.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/09/2016] [Accepted: 05/09/2016] [Indexed: 01/14/2023]
|
40
|
Curley JP, Champagne FA. Influence of maternal care on the developing brain: Mechanisms, temporal dynamics and sensitive periods. Front Neuroendocrinol 2016; 40:52-66. [PMID: 26616341 PMCID: PMC4783284 DOI: 10.1016/j.yfrne.2015.11.001] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/17/2015] [Accepted: 11/22/2015] [Indexed: 02/08/2023]
Abstract
Variation in maternal care can lead to divergent developmental trajectories in offspring with implications for neuroendocrine function and behavioral phenotypes. Study of the long-term outcomes associated with mother-infant interactions suggests complex mechanisms linking the experience of variation in maternal care and these neurobiological consequences. Through integration of genetic, molecular, cellular, neuroanatomical, and neuroendocrine approaches, significant advances in our understanding of these complex pathways have been achieved. In this review, we will consider the impact of maternal care on male and female offspring development with a particular focus on the issues of timing and mechanism. Identifying the period of sensitivity to maternal care and the temporal dynamics of the molecular and neuroendocrine changes that are a consequence of maternal care represents a critical step in the study of mechanism.
Collapse
Affiliation(s)
- James P Curley
- Department of Psychology, Columbia University, Room 406 Schermerhorn Hall, 1190 Amsterdam Avenue, New York, NY 10027, USA; Center for Integrative Animal Behavior, Columbia University, 1200 Amsterdam Avenue, New York, NY 10027, USA.
| | - Frances A Champagne
- Department of Psychology, Columbia University, Room 406 Schermerhorn Hall, 1190 Amsterdam Avenue, New York, NY 10027, USA; Center for Integrative Animal Behavior, Columbia University, 1200 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
41
|
Kentner AC. Neuroprotection and recovery from early-life adversity: considerations for environmental enrichment. Neural Regen Res 2015; 10:1545-7. [PMID: 26692834 PMCID: PMC4660730 DOI: 10.4103/1673-5374.165315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly Massachusetts College of Pharmacy & Health Sciences), Boston, MA, USA
| |
Collapse
|
42
|
Bolten M. Transgenerational Transmission of Stress Pathology. ZEITSCHRIFT FUR PSYCHOLOGIE-JOURNAL OF PSYCHOLOGY 2015. [DOI: 10.1027/2151-2604/a000219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract. The impact of the environment early in life on long-term outcomes is well known. Stressful experiences during pre- and postnatal development can modulate the genetic programming of specific brain circuits underlying emotional and cognitive aspects of behavioral adaptation to stressful experiences later in life. Furthermore, there is documented evidence for gene-environment interactions in the context of early-life stress. Identical gene variants can be associated with different phenotypes depending on environmental factors. DNA methylation, an enzymatically-catalyzed modification of the DNA, is the mechanism through which phenotypes are regulated. The dynamics and plasticity of epigenetic mechanisms can have short-term, long-term, or transgenerational consequences. In epigenetic research, rodent models have targeted several behavioral and emotional phenotypes. These models have contributed significantly to our understanding of the environmental regulation of the developmental brain in early life. This review will highlight studies with rats and mice on epigenetic processes in fetal programming of stress-related mental disorders.
Collapse
Affiliation(s)
- Margarete Bolten
- Child and Adolescent Psychiatric Clinic, University of Basel, Switzerland
| |
Collapse
|
43
|
Takatsuru Y, Koibuchi N. Alteration of somatosensory response in adulthood by early life stress. Front Mol Neurosci 2015; 8:15. [PMID: 26041988 PMCID: PMC4436820 DOI: 10.3389/fnmol.2015.00015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/05/2015] [Indexed: 01/01/2023] Open
Abstract
Early life stress is well-known as a critical risk factor for mental and cognitive disorders in adulthood. Such disorders are accompanied by altered neuro- (synapto-) genesis and gene expression. Because psychosomatic disorders induced by early life stress (e.g., physical and/or sexual abuse, and neglect) have become a socio-economic problem, it is very important to clarify the mechanisms underlying these changes. However, despite of intensive clinical and animal studies, such mechanisms have not yet been clarified. Although the disturbance of glucocorticoid and glutamate homeostasis by stress has been well-documented, it has not yet been clarified whether such disturbance by early life stress persists for life. Furthermore, since previous studies have focused on the detection of changes in specific brain regions, such as the hippocampus and prefrontal cortex, it has not been clarified whether early life stress induced changes in the sensory/motor system. Thus, in this review, we introduce recent studies on functional/structural changes in the somatosensory cortex induced by early life stress. We believe that this review provides new insights into the functional alteration of the somatosensory system induced by early life stress. Such information may have clinical relevance in terms of providing effective therapeutic interventions to early life stressed individuals.
Collapse
Affiliation(s)
- Yusuke Takatsuru
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi Japan
| |
Collapse
|
44
|
Tracing the trajectory of behavioral impairments and oxidative stress in an animal model of neonatal inflammation. Neuroscience 2015; 298:455-66. [PMID: 25934038 DOI: 10.1016/j.neuroscience.2015.04.048] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 11/21/2022]
Abstract
Exposure to early-life inflammation results in time-of-challenge-dependent changes in both brain and behavior. The consequences of this neural and behavioral reprogramming are most often reported in adulthood. However, the trajectory for the expression of these various changes is not well delineated, particularly between the juvenile and adult phases of development. Moreover, interventions to protect against these neurodevelopmental disruptions are rarely evaluated. Here, female Sprague-Dawley rats were housed in either environmental enrichment (EE) or standard care (SC) and their male and female offspring were administered 50 μg/kg i.p. of lipopolysaccharide (LPS) or pyrogen-free saline in a dual-administration neonatal protocol. All animals maintained their respective housing assignments from breeding until the end of the study. LPS exposure on postnatal days (P) 3 and 5 of life resulted in differential expression of emotional and cognitive disruptions and evidence of oxidative stress across development. Specifically, social behavior was reduced in neonatal-treated (n)LPS animals at adolescence (P40), but not adulthood (P70). In contrast, male nLPS rats exhibited intact spatial memory as adolescents which was impaired in later life. Moreover, these males had decreased prefrontal cortex levels of glutathione at P40, which was normalized in adult animals. Notably, EE appeared to offer some protection against the consequences of inflammation on juvenile social behavior and fully prevented reduced glutathione levels in the juvenile prefrontal cortex. Combined, these time-dependent effects provide evidence that early-life inflammation interacts with other developmental variables, specifically puberty and EE, in the expression (and prevention) of select behavioral and molecular programs.
Collapse
|