1
|
Kribakaran S, DeCross SN, Odriozola P, McLaughlin KA, Gee DG. Developmental Differences in a Hippocampal-Cingulate Pathway Involved in Learned Safety Following Interpersonal Trauma Exposure. J Am Acad Child Adolesc Psychiatry 2024:S0890-8567(24)01932-4. [PMID: 39368629 DOI: 10.1016/j.jaac.2024.07.928] [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: 12/26/2023] [Revised: 07/22/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
OBJECTIVE Nearly 65% of youth experience trauma, and up to one-third of youth with trauma exposure face profound mental health sequelae. There remains a need to elucidate factors that contribute to psychopathology following trauma exposure, and to optimize interventions for youth who do not benefit sufficiently from existing treatments. Here, we probe safety signal learning (SSL), which is a mechanism of fear reduction that leverages learned safety to inhibit fear in the presence of threat-associated stimuli and has been shown to attenuate fear via a hippocampal-cingulate--specifically, a dorsal anterior cingulate cortex (dACC)--pathway. METHOD The present study used behavioral and task-based functional magnetic resonance imaging data to examine age-related associations between interpersonal trauma exposure and the behavioral and neural correlates (ie, activation and functional connectivity) of SSL in a group of 102 youth (aged 9-19 years; 46 female, 56 male) with (n = 52) and without (n = 50) interpersonal trauma exposure. Primary analyses examined anterior hippocampal activation and anterior hippocampus-dACC functional connectivity. Exploratory analyses examined centromedial (CMA) and laterobasal (LBA) amygdala activation and anterior hippocampal, CMA, and LBA functional connectivity with additional anterior cingulate subregions (ie, subgenual anterior cingulate cortex [sgACC] and rostral anterior cingulate cortex [rosACC]). RESULTS Both youth with and without interpersonal trauma exposure successfully learned conditioned safety, which was determined by using self-report of contingency awareness. Youth with interpersonal trauma exposure (relative to youth in the comparison group) exhibited age-specific patterns of lower hippocampal activation (F2,96 = 3.75, pFDR = .049, ηp2 = 0.072), and, in exploratory analyses, showed heightened centromedial amygdala activation (F1,96 = 5.37, pFDR = .046, ηp2 = 0.053) and an age-related decrease in hippocampal-sgACC functional connectivity during SSL (F1,94 = 10.68, pFDR = .015, ηp2 = 0.102). We also show that hippocampal-sgACC functional connectivity mediated the association between interpersonal trauma exposure and post-traumatic stress disorder symptoms in an age-specific manner in the overall sample. CONCLUSION Together, these findings suggest that although age- and trauma-specific differences in the neural correlates of SSL may relate to the development of psychopathology, youth with interpersonal trauma exposure demonstrate successful learning of conditioned safety over time. DIVERSITY & INCLUSION STATEMENT We worked to ensure that the study questionnaires were prepared in an inclusive way. We worked to ensure sex and gender balance in the recruitment of human participants. We worked to ensure race, ethnic, and/or other types of diversity in the recruitment of human participants. While citing references scientificall`y relevant for this work, we also actively worked to promote sex and gender balance in our reference list. While citing references scientifically relevant for this work, we also actively worked to promote inclusion of historically underrepresented racial and/or ethnic groups in science in our reference list. We actively worked to promote sex and gender balance in our author group. We actively worked to promote inclusion of historically underrepresented racial and/or ethnic groups in science in our author group. One or more of the authors of this paper self-identifies as a member of one or more historically underrepresented racial and/or ethnic groups in science. One or more of the authors of this paper received support from a program designed to increase minority representation in science.
Collapse
Affiliation(s)
- Sahana Kribakaran
- Yale University, New Haven, Connecticut; Yale School of Medicine, New Haven, Connecticut
| | | | | | | | | |
Collapse
|
2
|
Chan D, Baker KD, Richardson R. The impact of chronic fluoxetine treatment in adolescence or adulthood on context fear memory and perineuronal nets. Dev Psychobiol 2024; 66:e22501. [PMID: 38807259 DOI: 10.1002/dev.22501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024]
Abstract
Selective serotonin reuptake inhibitors, such as fluoxetine (Prozac), are commonly prescribed pharmacotherapies for anxiety. Fluoxetine may be a useful adjunct because it can reduce the expression of learned fear in adult rodents. This effect is associated with altered expression of perineuronal nets (PNNs) in the amygdala and hippocampus, two brain regions that regulate fear. However, it is unknown whether fluoxetine has similar effects in adolescents. Here, we investigated the effect of fluoxetine exposure during adolescence or adulthood on context fear memory and PNNs in the basolateral amygdala (BLA), the CA1 subregion of the hippocampus, and the medial prefrontal cortex in rats. Fluoxetine impaired context fear memory in adults but not in adolescents. Further, fluoxetine increased the number of parvalbumin (PV)-expressing neurons surrounded by a PNN in the BLA and CA1, but not in the medial prefrontal cortex, at both ages. Contrary to previous reports, fluoxetine did not shift the percentage of PNNs toward non-PV cells in either the BLA or CA1 in the adults, or adolescents. These findings demonstrate that fluoxetine differentially affects fear memory in adolescent and adult rats but does not appear to have age-specific effects on PNNs.
Collapse
Affiliation(s)
- Diana Chan
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kathryn D Baker
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| | - Rick Richardson
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Premachandran H, Wilkin J, Arruda-Carvalho M. Minimizing Variability in Developmental Fear Studies in Mice: Toward Improved Replicability in the Field. Curr Protoc 2024; 4:e1040. [PMID: 38713136 DOI: 10.1002/cpz1.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
In rodents, the first weeks of postnatal life feature remarkable changes in fear memory acquisition, retention, extinction, and discrimination. Early development is also marked by profound changes in brain circuits underlying fear memory processing, with heightened sensitivity to environmental influences and stress, providing a powerful model to study the intersection between brain structure, function, and the impacts of stress. Nevertheless, difficulties related to breeding and housing young rodents, preweaning manipulations, and potential increased variability within that population pose considerable challenges to developmental fear research. Here we discuss several factors that may promote variability in studies examining fear conditioning in young rodents and provide recommendations to increase replicability. We focus primarily on experimental conditions, design, and analysis of rodent fear data, with an emphasis on mouse studies. The convergence of anatomical, synaptic, physiological, and behavioral changes during early life may increase variability, but careful practice and transparency in reporting may improve rigor and consensus in the field. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Hanista Premachandran
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- These authors contributed equally to this work
| | - Jennifer Wilkin
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- These authors contributed equally to this work
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Bevandić J, Chareyron LJ, Bachevalier J, Cacucci F, Genzel L, Newcombe NS, Vargha-Khadem F, Ólafsdóttir HF. Episodic memory development: Bridging animal and human research. Neuron 2024; 112:1060-1080. [PMID: 38359826 PMCID: PMC11129319 DOI: 10.1016/j.neuron.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024]
Abstract
Human episodic memory is not functionally evident until about 2 years of age and continues to develop into the school years. Behavioral studies have elucidated this developmental timeline and its constituent processes. In tandem, lesion and neurophysiological studies in non-human primates and rodents have identified key neural substrates and circuit mechanisms that may underlie episodic memory development. Despite this progress, collaborative efforts between psychologists and neuroscientists remain limited, hindering progress. Here, we seek to bridge human and non-human episodic memory development research by offering a comparative review of studies using humans, non-human primates, and rodents. We highlight critical theoretical and methodological issues that limit cross-fertilization and propose a common research framework, adaptable to different species, that may facilitate cross-species research endeavors.
Collapse
Affiliation(s)
- Juraj Bevandić
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Loïc J Chareyron
- Cognitive Neuroscience and Neuropsychiatry, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK; Laboratory of Brain and Cognitive Development, Institute of Psychology, University of Lausanne, Lausanne, Switzerland
| | - Jocelyne Bachevalier
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Department of Psychology, Emory University, Atlanta, GA, USA.
| | - Francesca Cacucci
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Lisa Genzel
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| | - Nora S Newcombe
- Department of Psychology, Temple University, Philadelphia, PA, USA.
| | - Faraneh Vargha-Khadem
- Cognitive Neuroscience and Neuropsychiatry, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - H Freyja Ólafsdóttir
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
5
|
Colombel N, Ferreira G, Sullivan RM, Coureaud G. Dynamic developmental changes in neurotransmitters supporting infant attachment learning. Neurosci Biobehav Rev 2023; 151:105249. [PMID: 37257712 PMCID: PMC10754360 DOI: 10.1016/j.neubiorev.2023.105249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023]
Abstract
Infant survival relies on rapid identification, remembering and behavioral responsiveness to caregivers' sensory cues. While neural circuits supporting infant attachment learning have largely remained elusive in children, use of invasive techniques has uncovered some of its features in rodents. During a 10-day sensitive period from birth, newborn rodents associate maternal odors with maternal pleasant or noxious thermo-tactile stimulation, which gives rise to a preference and approach behavior towards these odors, and blockade of avoidance learning. Here we review the neural circuitry supporting this neonatal odor learning, unique compared to adults, focusing specifically on the early roles of neurotransmitters such as glutamate, GABA (Gamma-AminoButyric Acid), serotonin, dopamine and norepinephrine, in the olfactory bulb, the anterior piriform cortex and amygdala. The review highlights the importance of deepening our knowledge of age-specific infant brain neurotransmitters and behavioral functioning that can be translated to improve the well-being of children during typical development and aid in treatment during atypical development in childhood clinical practice, and the care during rearing of domestic animals.
Collapse
Affiliation(s)
- Nina Colombel
- Ecole Normale Supérieure de Lyon, Lyon 1 Claude Bernard University, Lyon, France
| | - Guillaume Ferreira
- FoodCircus group, NutriNeuro Lab, INRAE 1286, Bordeaux University, Bordeaux, France
| | - Regina M Sullivan
- Emotional Brain Institute, The Nathan Kline Institute, Orangeburg, NY, USA; Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, USA
| | - Gérard Coureaud
- Sensory NeuroEthology Group, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Lyon 1 University, Jean-Monnet University, Bron, France.
| |
Collapse
|
6
|
The lifetime impact of stress on fear regulation and cortical function. Neuropharmacology 2023; 224:109367. [PMID: 36464208 DOI: 10.1016/j.neuropharm.2022.109367] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
A variety of stressful experiences can influence the ability to form and subsequently inhibit fear memory. While nonsocial stress can impact fear learning and memory throughout the lifespan, psychosocial stressors that involve negative social experiences or changes to the social environment have a disproportionately high impact during adolescence. Here, we review converging lines of evidence that suggest that development of prefrontal cortical circuitry necessary for both social experiences and fear learning is altered by stress exposure in a way that impacts both social and fear behaviors throughout the lifespan. Further, we suggest that psychosocial stress, through its impact on the prefrontal cortex, may be especially detrimental during early developmental periods characterized by higher sociability. This article is part of the Special Issue on 'Fear, Anxiety and PTSD'.
Collapse
|
7
|
Kitt ER, Odriozola P, Gee DG. Extinction Learning Across Development: Neurodevelopmental Changes and Implications for Pediatric Anxiety Disorders. Curr Top Behav Neurosci 2023; 64:237-256. [PMID: 37532964 DOI: 10.1007/7854_2023_430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Alterations in extinction learning relate to the development and maintenance of anxiety disorders across the lifespan. While exposure therapy, based on principles of extinction, can be highly effective for treating anxiety, many patients do not show sufficient improvement following treatment. In particular, evidence suggests that exposure therapy does not work sufficiently for up to 40% of children who receive this evidence-based treatment.Importantly, fear learning and extinction, as well as the neural circuitry supporting these processes, undergo dynamic changes across development. An improved understanding of developmental changes in extinction learning and the associated neural circuitry may help to identify targets to improve treatment response in clinically anxious children and adolescents. In this chapter, we provide a brief overview of methods used to study fear learning and extinction in developmental populations. We then review what is currently known about the developmental changes that occur in extinction learning and related neural circuitry. We end this chapter with a discussion of the implications of these neurodevelopmental changes for the characterization and treatment of pediatric anxiety disorders.
Collapse
Affiliation(s)
| | - Paola Odriozola
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Dylan G Gee
- Department of Psychology, Yale University, New Haven, CT, USA.
| |
Collapse
|
8
|
Singh S, Topolnik L. Inhibitory circuits in fear memory and fear-related disorders. Front Neural Circuits 2023; 17:1122314. [PMID: 37035504 PMCID: PMC10076544 DOI: 10.3389/fncir.2023.1122314] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/17/2023] [Indexed: 04/11/2023] Open
Abstract
Fear learning and memory rely on dynamic interactions between the excitatory and inhibitory neuronal populations that make up the prefrontal cortical, amygdala, and hippocampal circuits. Whereas inhibition of excitatory principal cells (PCs) by GABAergic neurons restrains their excitation, inhibition of GABAergic neurons promotes the excitation of PCs through a process called disinhibition. Specifically, GABAergic interneurons that express parvalbumin (PV+) and somatostatin (SOM+) provide inhibition to different subcellular domains of PCs, whereas those that express the vasoactive intestinal polypeptide (VIP+) facilitate disinhibition of PCs by inhibiting PV+ and SOM+ interneurons. Importantly, although the main connectivity motifs and the underlying network functions of PV+, SOM+, and VIP+ interneurons are replicated across cortical and limbic areas, these inhibitory populations play region-specific roles in fear learning and memory. Here, we provide an overview of the fear processing in the amygdala, hippocampus, and prefrontal cortex based on the evidence obtained in human and animal studies. Moreover, focusing on recent findings obtained using genetically defined imaging and intervention strategies, we discuss the population-specific functions of PV+, SOM+, and VIP+ interneurons in fear circuits. Last, we review current insights that integrate the region-specific inhibitory and disinhibitory network patterns into fear memory acquisition and fear-related disorders.
Collapse
Affiliation(s)
- Sanjay Singh
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
- *Correspondence: Lisa Topolnik
| |
Collapse
|
9
|
Awad W, Kritman M, Ferreira G, Maroun M. Differential Recruitment of the Infralimbic Cortex in Recent and Remote Retrieval and Extinction of Aversive Memory in Post-Weanling Rats. Int J Neuropsychopharmacol 2022; 25:489-497. [PMID: 35134947 PMCID: PMC9211009 DOI: 10.1093/ijnp/pyac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND We previously showed that the infralimbic medial prefrontal cortex (IL-mPFC) plays an important role in recent and remote memory retrieval and extinction of conditioned odor aversion (COA) and contextual fear conditioning (CFC) in adult rats. Because the mPFC undergoes maturation during post-weaning, here, we aimed to explore (1) whether post-weanling rats can form recent and remote COA and CFC memory, and (2) the role of the IL-mPFC in mediating these processes. METHODS To investigate the retrieval process, we transiently inactivated the IL-mPFC with lidocaine prior to the retrieval test at either recent or remote time points. To target the consolidation process, we applied the protein synthesis inhibitor after the retrieval at recent or remote time points. RESULTS Our results show that the post-weanling animals were able to develop both recent and remote memory of both COA and CFC. IL-mPFC manipulations had no effect on retrieval or extinction of recent and remote COA memory, suggesting that the IL has no effect in COA at this developmental stage. In contrast, the IL-mPFC played a role in (1) the extinction of recent, but not remote, CFC memory, and (2) the retrieval of remote, but not recent, CFC memory. Moreover, remote, but not recent, CFC retrieval enhanced c-Fos protein expression in the IL-mPFC. CONCLUSIONS Altogether, these results point to a differential role of the IL-mPFC in recent and remote CFC memory retrieval and extinction and further confirm the differences in the role of IL-mPFC in these processes in post-weanling and adult animals.
Collapse
Affiliation(s)
| | | | | | - Mouna Maroun
- Correspondence: Mouna Maroun, PhD, Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 31095, Israel ()
| |
Collapse
|
10
|
Roy N, Parhar I. Habenula orphan G-protein coupled receptors in the pathophysiology of fear and anxiety. Neurosci Biobehav Rev 2021; 132:870-883. [PMID: 34801259 DOI: 10.1016/j.neubiorev.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
The phasic emotion, fear, and the tonic emotion, anxiety, have been conventionally inspected in clinical frameworks to epitomize memory acquisition, storage, and retrieval. However, inappropriate expression of learned fear in a safe environment and its resistance to suppression is a cardinal feature of various fear-related disorders. A significant body of literature suggests the involvement of extra-amygdala circuitry in fear disorders. Consistent with this view, the present review underlies incentives for the association between the habenula and fear memory. G protein-coupled receptors (GPCRs) are important to understand the molecular mechanisms central to fear learning due to their neuromodulatory role. The efficacy of a pharmacological strategy aimed at exploiting habenular-GPCR desensitization machinery can serve as a therapeutic target combating the pathophysiology of fear disorders. Originating from this milieu, the conserved nature of orphan GPCRs in the brain, with some having the highest expression in the habenula can lead to recent endeavors in understanding its functionality in fear circuitry.
Collapse
Affiliation(s)
- Nisa Roy
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
11
|
Shan X, Contreras MP, Mendez M, Born J, Inostroza M. Unfolding of spatial representation at systems level in infant rats. Hippocampus 2021; 32:121-133. [PMID: 34786798 DOI: 10.1002/hipo.23392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 10/25/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022]
Abstract
Spatial representations enable navigation from early life on. However, the brain regions essential to form spatial representations, like the hippocampus, are considered functionally immature before weaning. Here, we examined the formation of representations of space in rat pups on postnatal day (PD) 16, using a simple habituation paradigm where the pups were exposed to an arena on three occasions, separated by ~140 min. Whereas on the first two occasions the arena was the same, on the third "test" occasion either proximal cues (Prox group), or distal cues (Dist group), or proximal and distal cues (Prox-Dist group), or no cues (No-change group) were rearranged. Locomotion (distance traveled) was used as behavioral measure of habituation, and c-Fos expression to measure regional brain activity at test. Locomotion generally decreased across the first two occasions. At test, it reached a minimum in the No-change group, indicating familiarity with the spatial conditions. By contrast, the Prox-Dist group displayed a significant increase in locomotion which was less robust in the Prox group and absent in the Dist group, a pattern suggesting that the pups relied more on proximal than distal cues during spatial exploration. c-Fos activity in the No-change group was significantly suppressed in the hippocampus (CA1, CA3, dentate gyrus) but simultaneously enhanced in the prelimbic area (PL) of the medial prefrontal cortex, compared with untreated Home-cage controls, pointing to a possible involvement of the PL in regulating locomotion in familiar spaces. By contrast, in both Prox-Dist and Prox groups c-Fos activity was enhanced in hippocampal CA1 and CA3 regions, suggesting these regions might be particularly involved in regulating exploration of spatial novelty. Our findings show that functional representations of space at a systems level are formed already in pre-weanling rats.
Collapse
Affiliation(s)
- Xia Shan
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,Graduate School of Neural & Behavioral Science, International Max Planck Research School, Tübingen, Germany
| | - María P Contreras
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,Graduate School of Neural & Behavioral Science, International Max Planck Research School, Tübingen, Germany
| | - Marta Mendez
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Plaza Feijoo, Oviedo, Spain
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD), Institute for Diabetes Research & Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Tübingen, Germany.,Werner Reichert Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Marion Inostroza
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Carstens KE, Lustberg DJ, Shaughnessy EK, McCann KE, Alexander GM, Dudek SM. Perineuronal net degradation rescues CA2 plasticity in a mouse model of Rett syndrome. J Clin Invest 2021; 131:e137221. [PMID: 34228646 DOI: 10.1172/jci137221] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
Perineuronal nets (PNNs), a specialized form of extracellular matrix, are abnormal in the brains of people with Rett syndrome (RTT). We previously reported that PNNs function to restrict synaptic plasticity in hippocampal area CA2, which is unusually resistant to long-term potentiation (LTP) and has been linked to social learning in mice. Here we report that PNNs appear elevated in area CA2 of the hippocampus of an individual with RTT and that PNNs develop precociously and remain elevated in area CA2 of a mouse model of RTT (Mecp2-null). Further, we provide evidence that LTP could be induced at CA2 synapses prior to PNN maturation (postnatal day 8-11) in wild-type mice and that this window of plasticity was prematurely restricted at CA2 synapses in Mecp2-null mice. Degrading PNNs in Mecp2-null hippocampus was sufficient to rescue the premature disruption of CA2 plasticity. We identified several molecular targets that were altered in the developing Mecp2-null hippocampus that may explain aberrant PNNs and CA2 plasticity, and we discovered that CA2 PNNs are negatively regulated by neuronal activity. Collectively, our findings demonstrate that CA2 PNN development is regulated by Mecp2 and identify a window of hippocampal plasticity that is disrupted in a mouse model of RTT.
Collapse
|
13
|
Osborne BF, Beamish SB, Schwarz JM. The effects of early-life immune activation on microglia-mediated neuronal remodeling and the associated ontogeny of hippocampal-dependent learning in juvenile rats. Brain Behav Immun 2021; 96:239-255. [PMID: 34126173 PMCID: PMC8319153 DOI: 10.1016/j.bbi.2021.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/11/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022] Open
Abstract
Many neurodevelopmental disorders and associated learning deficits have been linked to early-life immune activation or ongoing immune dysregulation (Laskaris et al., 2016; O'Connor et al., 2014; Frick et al., 2013). Neuroscientists have begun to understand how the maturation of neural circuits allows for the emergence of cognitive and learning behaviors; yet we know very little about how these developing neural circuits are perturbed by certain events, including risk-factors such as early-life immune activation and immune dysregulation. To answer these questions, we examined the impact of early-life immune activation on the emergence of hippocampal-dependent learning in juvenile male and female rats using a well-characterized hippocampal-dependent learning task and we investigated the corresponding, dynamic multicellular interactions in the hippocampus that may contribute to these learning deficits. We found that even low levels of immune activation can result in hippocampal-depedent learning deficits days later, but only when this activation occurs during a sensitive period of development. The initial immune response and associated cytokine production in the hippocampus resolved within 24 h, several days prior to the observed learning deficit, but notably the initial immune response was followed by altered microglial-neuronal communication and synapse remodeling that changed the structure of hippocampal neurons during this period of juvenile brain development. We conclude that immune activation or dysregulation during a sensitive period of hippocampal development can precipitate the emergence of learning deficits via a multi-cellular process that may be initiated by, but not the direct result of the initial cytokine response. SIGNIFICANCE STATEMENT: Many neurodevelopmental disorders have been linked to early-life immune activation or immune dysregulation; however, very little is known about how dynamic changes in neuroimmune cells mediate the transition from normal brain function to the early stages of cognitive disorders, or how changes in immune signaling are subsequently integrated into developing neuronal networks. The current experiments examined the consequences of immune activation on the cellular and molecular changes that accompany the emergence of learning deficits during a sensitive period of hippocampal development. These findings have the potential to significantly advance our understanding of how early-life immune activation or dysregulation can result in the emergence of cognitive and learning deficits that are the largest source of years lived with disability in humans.
Collapse
Affiliation(s)
- Brittany F. Osborne
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| | - Sarah B. Beamish
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| | - Jaclyn M. Schwarz
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| |
Collapse
|
14
|
Sullivan RM, Opendak M. Neurobiology of Infant Fear and Anxiety: Impacts of Delayed Amygdala Development and Attachment Figure Quality. Biol Psychiatry 2021; 89:641-650. [PMID: 33109337 PMCID: PMC7914291 DOI: 10.1016/j.biopsych.2020.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/16/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Anxiety disorders are the most common form of mental illness and are more likely to emerge during childhood compared with most other psychiatric disorders. While research on children is the gold standard for understanding the behavioral expression of anxiety and its neural circuitry, the ethical and technical limitations in exploring neural underpinnings limit our understanding of the child's developing brain. Instead, we must rely on animal models to build strong methodological bridges for bidirectional translation to child development research. Using the caregiver-infant context, we review the rodent literature on early-life fear development to characterize developmental transitions in amygdala function underlying age-specific behavioral transitions. We then describe how this system can be perturbed by early-life adversity, including reduced efficacy of the caregiver as a safe haven. We suggest that greater integration of clinically informed animal research enhances bidirectional translation to permit new approaches to therapeutics for children with early onset anxiety disorders.
Collapse
Affiliation(s)
- Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute, New York, NY USA,Child & Adolescent Psychiatry, New York University Langone Medical Center, New York, NY USA
| | - Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute, New York, NY USA,Child & Adolescent Psychiatry, New York University Langone Medical Center, New York, NY USA
| |
Collapse
|
15
|
Phasuk S, Pairojana T, Suresh P, Yang CH, Roytrakul S, Huang SP, Chen CC, Pakaprot N, Chompoopong S, Nudmamud-Thanoi S, Liu IY. Enhanced contextual fear memory in peroxiredoxin 6 knockout mice is associated with hyperactivation of MAPK signaling pathway. Mol Brain 2021; 14:42. [PMID: 33632301 PMCID: PMC7908735 DOI: 10.1186/s13041-021-00754-1] [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: 12/12/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Fear dysregulation is one of the symptoms found in post-traumatic stress disorder (PTSD) patients. The functional abnormality of the hippocampus is known to be implicated in the development of such pathology. Peroxiredoxin 6 (PRDX6) belongs to the peroxiredoxin family. This antioxidant enzyme is expressed throughout the brain, including the hippocampus. Recent evidence reveals that PRDX6 plays an important role in redox regulation and the modulation of several signaling molecules involved in fear regulation. Thus, we hypothesized that PRDX6 plays a role in the regulation of fear memory. We subjected a systemic Prdx6 knockout (Prdx6-/-) mice to trace fear conditioning and observed enhanced fear response after training. Intraventricular injection of lentivirus-carried mouse Prdx6 into the 3rd ventricle reduced the enhanced fear response in these knockout mice. Proteomic analysis followed by validation of western blot analysis revealed that several proteins in the MAPK pathway, such as NTRK2, AKT, and phospho-ERK1/2, cPLA2 were significantly upregulated in the hippocampus of Prdx6-/- mice during the retrieval stage of contextual fear memory. The distribution of PRDX6 found in the astrocytes was also observed throughout the hippocampus. This study identifies PRDX6 as a participant in the regulation of fear response. It suggests that PRDX6 and related molecules may have important implications for understanding fear-dysregulation associated disorders like PTSD.
Collapse
Affiliation(s)
- Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tanita Pairojana
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Pavithra Suresh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Chee-Hing Yang
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supin Chompoopong
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sutisa Nudmamud-Thanoi
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Ingrid Y. Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
16
|
Stanton ME, Murawski NJ, Jablonski SA, Robinson-Drummer PA, Heroux NA. Mechanisms of context conditioning in the developing rat. Neurobiol Learn Mem 2021; 179:107388. [PMID: 33482320 DOI: 10.1016/j.nlm.2021.107388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
The article reviews our studies of contextual fear conditioning (CFC) in rats during a period of development---Postnatal Day (PND) 17-33---that represents the late-infant, juvenile, and early-adolescent stages. These studies seek to acquire 'systems level' knowledge of brain and memory development and apply it to a rodent model of Fetal Alcohol Spectrum Disorder (FASD). This rodent model focuses on alcohol exposure from PND4-9, a period of brain development equivalent to the human third trimester, when neocortex, hippocampus, and cerebellum are especially vulnerable to adverse effects of alcohol. Our research emphasizes a variant of CFC, termed the Context Preexposure Facilitation Effect (CPFE, Fanselow, 1990), in which context representations incidentally learned on one occasion are retrieved and associated with immediate shock on a subsequent occasion. These representations can be encoded at the earliest developmental stage but seem not to be retained or retrieved until the juvenile period. This is associated with developmental differences in context-elicited expression, in prefrontal cortex, hippocampus, and amygdala, of immediate early genes (IEGs) that are implicated in long-term memory. Loss-of-function studies establish a functional role for these regions as soon as the CPFE emerges during ontogeny. In our rodent model of FASD, the CPFE is much more sensitive to alcohol dose than other commonly used cognitive tasks. This impairment can be reversed by acute administration during behavioral testing of drugs that enhance cholinergic function. This effect is associated with normalized IEG expression in prefrontal cortex during incidental context learning. In summary, our findings suggest that long-term memory of incidentally-learned context representations depends on prefrontal-hippocampal circuitry that is important both for the normative development of context conditioning and for its disruption by developmental alcohol exposure.
Collapse
Affiliation(s)
- Mark E Stanton
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - Nathen J Murawski
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - Sarah A Jablonski
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | | | - Nicholas A Heroux
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| |
Collapse
|
17
|
Mooney LN, Johnson EG, Prabhakar J, Ghetti S. Memory-related hippocampal activation during sleep and temporal memory in toddlers. Dev Cogn Neurosci 2020; 47:100908. [PMID: 33395613 PMCID: PMC7785886 DOI: 10.1016/j.dcn.2020.100908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 11/12/2022] Open
Abstract
Two-year-olds played tablet games requiring them to remember locations visited by characters and the temporal order with which they visited them. Two-year-olds remembered spatial locations better than temporal order immediately after learning, and after a 20-minute and one-week delay. Song-related hippocampal activation during sleep is associated with memory for temporal order.
Nonhuman research has implicated developmental processes within the hippocampus in the emergence and early development of episodic memory, but research in humans has been constrained by the difficulty of examining hippocampal function during early development. In the present study, we assessed 48 2-year-olds with a novel paradigm in which participants completed two games on a tablet that required remembering associations between unique characters, the places they visited, and the temporal order with which they did so. At the completion of each game, a unique, novel song played. Toddlers remembered spatial locations better than temporal order during an immediate test, after a 20-minute delay, and after a week delay. After the last behavioral session, toddlers underwent an fMRI task during natural nocturnal sleep evaluating hippocampal activation in response to learned and novel songs. We found that the extent of hippocampal activation for learned songs compared to novel songs during sleep was correlated with memory for temproal order across all time delays, but not with memory for spatial locations. The results confirm that that the functional contribution of the hippocampus to early memory can be assessed during sleep and suggest that assessment of temporal aspects of memory in the current task best capture this contribution.
Collapse
Affiliation(s)
- Lindsey N Mooney
- Center for Mind and Brain, University of California, Davis, United States; Department of Psychology, University of California, Davis, United States.
| | - Elliott G Johnson
- Center for Mind and Brain, University of California, Davis, United States; Human Development Graduate Group, University of California, Davis, United States
| | - Janani Prabhakar
- Center for Mind and Brain, University of California, Davis, United States
| | - Simona Ghetti
- Center for Mind and Brain, University of California, Davis, United States; Human Development Graduate Group, University of California, Davis, United States; Department of Psychology, University of California, Davis, United States.
| |
Collapse
|
18
|
Kim J, Lee S, Kim J, Ham S, Park JHY, Han S, Jung YK, Shim I, Han JS, Lee KW, Kim J. Ca2+-permeable TRPV1 pain receptor knockout rescues memory deficits and reduces amyloid-β and tau in a mouse model of Alzheimer's disease. Hum Mol Genet 2020; 29:228-237. [PMID: 31814000 DOI: 10.1093/hmg/ddz276] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/14/2019] [Accepted: 11/11/2019] [Indexed: 01/31/2023] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) protein is a pain receptor that elicits a hot sensation when an organism eats the capsaicin of red chili peppers. This calcium (Ca2+)-permeable cation channel is mostly expressed in the peripheral nervous system sensory neurons but also in the central nervous system (e.g. hippocampus and cortex). Preclinical studies found that TRPV1 mediates behaviors associated with anxiety and depression. Loss of TRPV1 functionality increases expression of genes related to synaptic plasticity and neurogenesis. Thus, we hypothesized that TRPV1 deficiency may modulate Alzheimer's disease (AD). We generated a triple-transgenic AD mouse model (3xTg-AD+/+) with wild-type (TRPV1+/+), hetero (TRPV1+/-) and knockout (TRPV1-/-) TRPV1 to investigate the role of TRPV1 in AD pathogenesis. We analyzed the animals' memory function, hippocampal Ca2+ levels and amyloid-β (Aβ) and tau pathologies when they were 12 months old. We found that compared with 3xTg-AD-/-/TRPV1+/+ mice, 3xTg-AD+/+/TRPV1+/+ mice had memory impairment and increased levels of hippocampal Ca2+, Aβ and total and phosphorylated tau. However, 3xTg-AD+/+/TRPV1-/- mice had better memory function and lower levels of hippocampal Ca2+, Aβ, tau and p-tau, compared with 3xTg-AD+/+/TRPV1+/+ mice. Examination of 3xTg-AD-derived primary neuronal cultures revealed that the intracellular Ca2+ chelator BAPTA/AM and the TRPV1 antagonist capsazepine decreased the production of Aβ, tau and p-tau. Taken together, these results suggested that TRPV1 deficiency had anti-AD effects and promoted resilience to memory loss. These findings suggest that drugs or food components that modulate TRPV1 could be exploited as therapeutics to prevent or treat AD.
Collapse
Affiliation(s)
- Juyong Kim
- Department of Agricultural Biotechnology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Siyoung Lee
- Department of Agricultural Biotechnology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jaekyoon Kim
- Department of Agricultural Biotechnology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Sangwoo Ham
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jung Han Yoon Park
- Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seungbong Han
- Department of Applied Statistics, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jung-Soo Han
- Department of Biological Sciences, Konkuk University, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea.,Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jiyoung Kim
- Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
| |
Collapse
|
19
|
Konrad C, Adolph D, Herbert JS, Neuhoff L, Mohr C, Jagusch-Poirier J, Seehagen S, Weigelt S, Schneider S. A New 3-Day Standardized Eyeblink Conditioning Protocol to Assess Extinction Learning From Infancy to Adulthood. Front Behav Neurosci 2020; 14:135. [PMID: 32922270 PMCID: PMC7457038 DOI: 10.3389/fnbeh.2020.00135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/13/2020] [Indexed: 11/25/2022] Open
Abstract
Associative learning can be observed from the neonatal period onward, providing opportunities to examine changes in basic learning and memory abilities. One method that is suitable to study associative learning is classical eyeblink conditioning (EBC) which is dependent on the cerebellum. Extinction learning can be systematically investigated in this paradigm by varying the context during learning and extinction. Because of methodological difficulties and ethical challenges, no studies have compared extinction learning using EBC across human development. Our goal was to test feasibility of a 3-day delay EBC paradigm that can be used from infancy to adulthood. Acceptance/safety was tested especially for infancy by investigating attrition rates and parental report on infant wellbeing. On a paradigm side, we tested if the paradigm leads to successful acquisition and extinction. An air puff served as unconditional stimulus (US) and a tone as conditional stimulus (CS). On day 1 during acquisition, participants received 36 US–CS pairings in context A. On day 2, participants received 12 acquisition trials in context A to consolidate association learning, followed by 48 extinction trials (tone alone presentations) in context B. Renewal was assessed on day 3 and incorporated 12 CS alone trials presented in both the acquisition context and the extinction context. Eyeblink responses were videotaped and coded offline. The protocol was tested with 12–36-months-old infants (N = 72), adolescents (N = 8), and adults (N = 8). Concerning the acceptance/safety side, attrition ranged from 21 to 58% in infant samples due to the complex preparation of the children for the paradigm. However, attrition is equal to or lower than other infant learning paradigms. Parents of infant samples were very interested in the paradigm and reported low levels of infant stress, exhaustion, and negative feelings during the sessions. Data quality was very high, and no participant had to be excluded because of insufficient data. Concerning the paradigm side, participants showed successful acquisition and extinction as a group. The procedure is ethically sound, feasible, tolerated by many infants, and acceptable among parents. The data show successful acquisition and extinction rates, making the paradigm a valuable tool for investigating developmental changes in extinction learning over the lifespan.
Collapse
Affiliation(s)
- Carolin Konrad
- Faculty of Psychology, Clinical Child and Adolescent Psychology, Mental Health Research and Treatment Center, Ruhr University Bochum, Bochum, Germany
| | - Dirk Adolph
- Faculty of Psychology, Clinical Child and Adolescent Psychology, Mental Health Research and Treatment Center, Ruhr University Bochum, Bochum, Germany
| | - Jane S Herbert
- Wollongong Infant Learning Lab, School of Psychology and Early Start, University of Wollongong, Wollongong, NSW, Australia
| | - Lina Neuhoff
- Faculty of Psychology, Clinical Child and Adolescent Psychology, Mental Health Research and Treatment Center, Ruhr University Bochum, Bochum, Germany
| | - Cornelia Mohr
- Abteilung für Kinderschutz, Vestische Kinder- und Jugendklinik Datteln, Universität Witten/Herdecke, Datteln, Germany
| | - Julie Jagusch-Poirier
- Vision, Visual Impairments & Blindness, Faculty of Rehabilitation Sciences, Technical University, Dortmund University, Dortmund, Germany
| | - Sabine Seehagen
- Developmental Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Sarah Weigelt
- Vision, Visual Impairments & Blindness, Faculty of Rehabilitation Sciences, Technical University, Dortmund University, Dortmund, Germany
| | - Silvia Schneider
- Faculty of Psychology, Clinical Child and Adolescent Psychology, Mental Health Research and Treatment Center, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
20
|
Xia D, Min C, Chen Y, Ling R, Chen M, Li X. Repetitive Pain in Neonatal Male Rats Impairs Hippocampus-Dependent Fear Memory Later in Life. Front Neurosci 2020; 14:722. [PMID: 32733201 PMCID: PMC7360690 DOI: 10.3389/fnins.2020.00722] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/17/2020] [Indexed: 11/13/2022] Open
Abstract
Preterm infants in neonatal intensive care units are inevitably subjected to numerous painful procedures. However, little is known about the consequences of early pain experience on fear memory formation later in life. We hypothesized that exposure to repetitive pain in early life triggered hippocampal synaptic plasticity and resulted in memory deficiency in prepubertal and adult rats. From the day of birth (P0) to postnatal day 7 (P7), neonatal male rat pups were randomly assigned to either needle pricks or tactile touches repetitively every 6 h. Trace fear conditioning was performed on rats on P24-P26 and P87-P89. On P24 and P87, rats were sacrificed for molecular and electrophysiological studies. On P24-26 and P87-89, rats that experienced neonatal needle treatment showed a significant reduction in freezing time in the contextual fear conditioning (P < 0.05) and trace fear conditioning tests (P < 0.05). Moreover, repetitive neonatal procedural pain caused a significant decrease in the magnitude of hippocampal long-term potentiation induced by high-frequency stimulation. Furthermore, rats that experienced neonatal needle treatment demonstrated sustained downregulation of NR1, NR2A, NR2B, and GluR1 expression in the hippocampus. Therefore, neonatal pain is related to deficits in hippocampus-related fear memory later in life and might be caused by impairments in hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Dongqing Xia
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Cuiting Min
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yinhua Chen
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ru Ling
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mengying Chen
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Investigation of GluA1 and GluA2 AMPA receptor subtype distribution in the hippocampus and anterior cingulate cortex of Long Evans rats during development. IBRO Rep 2020; 8:91-100. [PMID: 32300670 PMCID: PMC7152689 DOI: 10.1016/j.ibror.2020.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/22/2020] [Indexed: 11/21/2022] Open
Abstract
Preadolescent development is characterized by a reorganization of connectivity within and between brain regions that coincides with the emergence of complex behaviors. During the preadolescent period, the rodent hippocampus and regions of the frontal cortex are remodelled as the brain strengthens active connections and eliminates others. In the developing and mature brain, changes in the properties of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAr)-mediated synaptic responses contribute to experience-dependent changes in neural organization and function. AMPAr are made up of 4 subunits, of which GluA1 and GluA2 have been shown to play the most prominent role in functional plasticity. In this study, we sought to determine whether levels of these two subunits changed during the course of pre-adolescent development in the hippocampus and anterior cingulate cortex (ACC). To investigate the developmental changes in GluA1 and GluA2 AMPAr subunits, Western blotting and immunohistochemistry were performed on the ACC and hippocampus from P18 - P30 and compared to adult (P50) levels and distribution. Within the hippocampus, protein levels of GluA1 and GluA2 peaked around P26-30 whereby localized staining in the dentate gyrus reflected this pattern. GluA1 and GluA2 levels within the ACC showed little variation during this developmental period. These results indicate that changes in AMPAr subunits within the hippocampus coincide with developmental modifications that underlie the shift from juvenile- to adult-like capabilities. However, changes in AMPAr distribution in the ACC might not mediate changes that reflect preadolescent developmental shifts.
Collapse
|
22
|
Lander SS, Chornyy S, Safory H, Gross A, Wolosker H, Gaisler‐Salomon I. Glutamate dehydrogenase deficiency disrupts glutamate homeostasis in hippocampus and prefrontal cortex and impairs recognition memory. GENES BRAIN AND BEHAVIOR 2020; 19:e12636. [DOI: 10.1111/gbb.12636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/11/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Affiliation(s)
| | - Sergiy Chornyy
- Department of PsychologyUniversity of Haifa Haifa Israel
| | - Hazem Safory
- Department of Biochemistry, The Ruth and Bruce Rappaport Faculty of MedicineTechnion‐Israel Institute of Technology Haifa Israel
| | - Amit Gross
- Department of PsychologyUniversity of Haifa Haifa Israel
| | - Herman Wolosker
- Department of Biochemistry, The Ruth and Bruce Rappaport Faculty of MedicineTechnion‐Israel Institute of Technology Haifa Israel
| | | |
Collapse
|
23
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
24
|
During infant maltreatment, stress targets hippocampus, but stress with mother present targets amygdala and social behavior. Proc Natl Acad Sci U S A 2019; 116:22821-22832. [PMID: 31636210 DOI: 10.1073/pnas.1907170116] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Infant maltreatment increases vulnerability to physical and mental disorders, yet specific mechanisms embedded within this complex infant experience that induce this vulnerability remain elusive. To define critical features of maltreatment-induced vulnerability, rat pups were reared from postnatal day 8 (PN8) with a maltreating mother, which produced amygdala and hippocampal deficits and decreased social behavior at PN13. Next, we deconstructed the maltreatment experience to reveal sufficient and necessary conditions to induce this phenotype. Social behavior and amygdala deficits (volume, neurogenesis, c-Fos, local field potential) required combined chronic high corticosterone and maternal presence (not maternal behavior). Hippocampal deficits were induced by chronic high corticosterone regardless of social context. Causation was shown by blocking corticosterone during maltreatment and suppressing amygdala activity during social behavior testing. These results highlight (1) that early life maltreatment initiates multiple pathways to pathology, each with distinct causal mechanisms and outcomes, and (2) the importance of social presence on brain development.
Collapse
|
25
|
Ciric T, Cahill SP, Snyder JS. Dentate gyrus neurons that are born at the peak of development, but not before or after, die in adulthood. Brain Behav 2019; 9:e01435. [PMID: 31576673 PMCID: PMC6790299 DOI: 10.1002/brb3.1435] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/14/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION In the dentate gyrus of the rodent hippocampus, neurogenesis begins prenatally and continues to the end of life. Adult-born neurons often die in the first few weeks after mitosis, but those that survive to 1 month persist indefinitely. In contrast, neurons born at the peak of development are initially stable but can die later in adulthood. Physiological and pathological changes in the hippocampus may therefore result from both the addition of new neurons and the loss of older neurons. The extent of neuronal loss remains unclear since no studies have examined whether neurons born at other stages of development also undergo delayed cell death. METHODS We used BrdU to label dentate granule cells that were born in male rats on embryonic day 19 (E19; before the developmental peak), postnatal day 6 (P6; peak), and P21 (after the peak). We quantified BrdU+ neurons in separate groups of rats at 2 and 6 months post-BrdU injection to estimate cell death in young adulthood. RESULTS Consistent with previous work, there was a 15% loss of P6-born neurons between 2 and 6 months of age. In contrast, E19- or P21-born neurons were stable throughout young adulthood. DISCUSSION Delayed death of P6-born neurons suggests these cells may play a unique role in hippocampal plasticity adulthood, for example, by contributing to the turnover of hippocampal memory. Their loss may also play a role in disorders that are characterized by hippocampal atrophy.
Collapse
Affiliation(s)
- Tina Ciric
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Shaina P Cahill
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Holahan MR, Tzakis N, Oliveira FA. Developmental Aspects of Glucose and Calcium Availability on the Persistence of Memory Function Over the Lifespan. Front Aging Neurosci 2019; 11:253. [PMID: 31572169 PMCID: PMC6749050 DOI: 10.3389/fnagi.2019.00253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/27/2019] [Indexed: 01/09/2023] Open
Abstract
An important aspect concerning the underlying nature of memory function is an understanding of how memories are acquired and lost. The stability, and ultimate demise, of memory over the lifespan of an organism remains a critical topic in determining the neurobiological mechanisms that mediate memory representations. This has important implications for the elucidation and treatment of neurodegenerative diseases such as Alzheimer's disease (AD). One important question in the context of preserving functional plasticity over the lifespan is the determination of the neurobiological structural and functional changes that contribute to the formation of memory during the juvenile time frame that might provide protection against later memory dysfunction by promoting the establishment of redundant neural pathways. The main question being, if memory formation during the juvenile period does strengthen and preserve memory stability over the lifespan, what are the neurobiological structural or functional substrates that mediate this effect? One neural attribute whose function may be altered with early life experience and provide a mechanism to preserve memory through the lifespan is glucose transport-linked calcium (Ca2+) buffering. Because peak increases in glucose utilization overlap with a timeframe during which spatial training can enhance later memory processing, it might be the case that learning-associated changes in glucose utilization would provide an important neural functional change to preserve memory function throughout the lifespan. The glucose transporters are proteins that are reduced in AD pathology and there is evidence that glucose reductions can impair Ca2+ buffering. In the absence of an appropriate supply of ATP, provided via glucose transport and glycolysis, Ca2+ levels can rise leading to neural vulnerability with ensuing pathological outcomes. In this review, we explore the hypothesis that enhancing glucose utilization with spatial training during the preadolescent period will provide a functional enhancement that regulates glucose-dependent Ca2+ signaling during aging or neurodegeneration and provide essential neural resources to preserve functional plasticity and memory function.
Collapse
Affiliation(s)
- Matthew R. Holahan
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Laboratory of Cellular and Molecular Neurobiology (LaNeC), Center for Mathematics, Computing and Cognition, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Niko Tzakis
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Fernando A. Oliveira
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Laboratory of Cellular and Molecular Neurobiology (LaNeC), Center for Mathematics, Computing and Cognition, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| |
Collapse
|
27
|
Junod A, Opendak M, LeDoux JE, Sullivan RM. Development of Threat Expression Following Infant Maltreatment: Infant and Adult Enhancement but Adolescent Attenuation. Front Behav Neurosci 2019; 13:130. [PMID: 31293397 PMCID: PMC6603125 DOI: 10.3389/fnbeh.2019.00130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Early life maltreatment by the caregiver constitutes a major risk factor for the development of later-life psychopathologies, including fear-related pathologies. Here, we used an animal model of early life maltreatment induced by the Scarcity-Adversity Model of low bedding (LB) where the mother is given insufficient bedding for nest building while rat pups were postnatal days (PN) 8-12. To assess effects of maltreatment on the expression of threat-elicited defensive behaviors, animals underwent odor-shock threat conditioning at three developmental stages: late infancy (PN18), adolescence (PN45) or adulthood (>PN75) and tested the next day with odor only presentations (cue test). Results showed that in typically developing rats, the response to threat increases with maturation, although experience with maltreatment in early infancy produced enhanced responding to threat in infancy and adulthood, but a decrease in maltreated adolescents. To better understand the unique features of this decreased threat responding in adolescence, c-Fos expression was assessed within the amygdala and ventromedial prefrontal cortex (vmPFC) associated with the cued expression of threat learning. Fos counts across amygdala subregions were lower in LB rats compared to controls, while enhanced c-Fos expression was observed in the vmPFC prelimbic cortex (PL). Correlational analysis between freezing behavior and Fos revealed freezing levels were correlated with CeA in controls, although more global correlations were detected in LB-reared rats, including the BA, LA, and CeA. Functional connectivity analysis between brain regions showed that LB reared rats exhibited more diffuse interconnectivity across amygdala subnuclei, compared the more heterogeneous patterns observed in controls. In addition, functional connectivity between the IL and LA switched from positive to negative in abused adolescents. Overall, these results suggest that in adolescence, the unique developmental decrease in fear expression following trauma is associated with distinct changes in regional function and long-range connectivity, reminiscent of pathological brain function. These results suggest that early life maltreatment from the caregiver perturbs the developmental trajectory of threat-elicited behavior. Indeed, it is possible that this form of trauma, where the infant's safety signal or "safe haven" (the caregiver) is actually the source of the threat, produces distinct outcomes across development.
Collapse
Affiliation(s)
- Anouchka Junod
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Joseph E. LeDoux
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| |
Collapse
|
28
|
Perry RE, Finegood ED, Braren SH, DeJoseph ML, Putrino DF, Wilson DA, Sullivan RM, Raver CC, Blair C. Developing a neurobehavioral animal model of poverty: Drawing cross-species connections between environments of scarcity-adversity, parenting quality, and infant outcome. Dev Psychopathol 2019; 31:399-418. [PMID: 29606185 PMCID: PMC6168440 DOI: 10.1017/s095457941800007x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Children reared in impoverished environments are at risk for enduring psychological and physical health problems. Mechanisms by which poverty affects development, however, remain unclear. To explore one potential mechanism of poverty's impact on social-emotional and cognitive development, an experimental examination of a rodent model of scarcity-adversity was conducted and compared to results from a longitudinal study of human infants and families followed from birth (N = 1,292) who faced high levels of poverty-related scarcity-adversity. Cross-species results supported the hypothesis that altered caregiving is one pathway by which poverty adversely impacts development. Rodent mothers assigned to the scarcity-adversity condition exhibited decreased sensitive parenting and increased negative parenting relative to mothers assigned to the control condition. Furthermore, scarcity-adversity reared pups exhibited decreased developmental competence as indicated by disrupted nipple attachment, distress vocalization when in physical contact with an anesthetized mother, and reduced preference for maternal odor with corresponding changes in brain activation. Human results indicated that scarcity-adversity was inversely correlated with sensitive parenting and positively correlated with negative parenting, and that parenting fully mediated the association of poverty-related risk with infant indicators of developmental competence. Findings are discussed from the perspective of the usefulness of bidirectional-translational research to inform interventions for at-risk families.
Collapse
Affiliation(s)
| | | | | | | | - David F. Putrino
- Department of Telemedicine and Virtual Rehabilitation, Burke Medical Research Institute & Department of Rehabilitation Medicine, Weill Cornell Medicine
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute & Department of Child and Adolescent Psychiatry, New York University School of Medicine
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute & Department of Child and Adolescent Psychiatry, New York University School of Medicine
| | | | - Clancy Blair
- Department of Applied Psychology, New York University
| | | |
Collapse
|
29
|
Cowan CSM, Stylianakis AA, Richardson R. Early-life stress, microbiota, and brain development: probiotics reverse the effects of maternal separation on neural circuits underpinning fear expression and extinction in infant rats. Dev Cogn Neurosci 2019; 37:100627. [PMID: 30981894 PMCID: PMC6969299 DOI: 10.1016/j.dcn.2019.100627] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/17/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022] Open
Abstract
Early-life stress has pervasive, typically detrimental, effects on physical and mental health across the lifespan. In rats, maternal-separation stress results in premature expression of an adult-like profile of fear regulation that predisposes stressed rats to persistent fear, one of the hallmarks of clinical anxiety. Probiotic treatment attenuates the effects of maternal separation on fear regulation. However, the neural pathways underlying these behavioral changes are unknown. Here, we examined the neural correlates of stress-induced alterations in fear behavior and their reversal by probiotic treatment. Male Sprague-Dawley rats were exposed to either standard rearing conditions or maternal-separation stress (postnatal days [P] 2–14). Some maternally-separated (MS) animals were also exposed to probiotics (Lactobacillus rhamnosus and L. helveticus) via the maternal drinking water during the period of stress. Using immunohistochemistry, we demonstrated that stressed rat pups prematurely exhibit adult-like engagement of the medial prefrontal cortex during fear regulation, an effect that can be prevented using a probiotic treatment. The present results add to the cross-species evidence that early adversity hastens maturation in emotion-related brain circuits. Importantly, our results also demonstrate that the precocious neural maturation in stressed infants is prevented by a non-invasive probiotic treatment.
Collapse
Affiliation(s)
- Caitlin S M Cowan
- School of Psychology, The University of New South Wales, Sydney, Australia.
| | | | - Rick Richardson
- School of Psychology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
30
|
Opendak M, Sullivan RM. Unique infant neurobiology produces distinctive trauma processing. Dev Cogn Neurosci 2019; 36:100637. [PMID: 30889546 PMCID: PMC6969239 DOI: 10.1016/j.dcn.2019.100637] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/11/2018] [Accepted: 03/07/2019] [Indexed: 12/31/2022] Open
Abstract
Trauma experienced in early life has unique neurobehavioral outcomes related to later life psychiatric sequelae. Recent evidence has further highlighted the context of infant trauma as critical, with trauma experienced within species-atypical aberrations in caregiving quality as particularly detrimental. Using data from primarily rodent models, we review the literature on the interaction between trauma and attachment in early life, which highlights the role of the caregiver's presence in engagement of attachment brain circuitry and suppressing threat processing by the amygdala. Together these data suggest that infant trauma processing and its enduring effects are impacted by both the immaturity of brain areas for processing trauma and the unique functioning of the early-life brain, which is biased towards forming robust attachments regardless of the quality of care. Understanding the critical role of the caregiver in further altering early life brain processing of trauma is important for developing age-relevant treatment and interventions.
Collapse
Affiliation(s)
- Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA.
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA
| |
Collapse
|
31
|
Callaghan B, Meyer H, Opendak M, Van Tieghem M, Harmon C, Li A, Lee FS, Sullivan RM, Tottenham N. Using a Developmental Ecology Framework to Align Fear Neurobiology Across Species. Annu Rev Clin Psychol 2019; 15:345-369. [PMID: 30786246 DOI: 10.1146/annurev-clinpsy-050718-095727] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Children's development is largely dependent on caregiving; when caregiving is disrupted, children are at increased risk for numerous poor outcomes, in particular psychopathology. Therefore, determining how caregivers regulate children's affective neurobiology is essential for understanding psychopathology etiology and prevention. Much of the research on affective functioning uses fear learning to map maturation trajectories, with both rodent and human studies contributing knowledge. Nonetheless, as no standard framework exists through which to interpret developmental effects across species, research often remains siloed, thus contributing to the current therapeutic impasse. Here, we propose a developmental ecology framework that attempts to understand fear in the ecological context of the child: their relationship with their parent. By referring to developmental goals that are shared across species (to attach to, then, ultimately, separate from the parent), this framework provides a common grounding from which fear systems and their dysfunction can be understood, thus advancing research on psychopathologies and their treatment.
Collapse
Affiliation(s)
- Bridget Callaghan
- Department of Psychology, Columbia University, New York, NY 10027, USA; , , , .,Department of Psychiatry, Melbourne University, Melbourne, Victoria 3010, Australia
| | - Heidi Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; , ,
| | - Maya Opendak
- Department of Child and Adolescent Psychiatry, New York University, Langone Medical Center, New York, NY 10016, USA; .,Nathan S. Klein Institute for Psychiatric Research, Orangeburg, New York 10962, USA;
| | | | - Chelsea Harmon
- Department of Psychology, Columbia University, New York, NY 10027, USA; , , ,
| | - Anfei Li
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; , ,
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; , ,
| | - Regina M Sullivan
- Department of Child and Adolescent Psychiatry, New York University, Langone Medical Center, New York, NY 10016, USA; .,Nathan S. Klein Institute for Psychiatric Research, Orangeburg, New York 10962, USA;
| | - Nim Tottenham
- Department of Psychology, Columbia University, New York, NY 10027, USA; , , ,
| |
Collapse
|
32
|
Gee DG, Bath KG, Johnson CM, Meyer HC, Murty VP, van den Bos W, Hartley CA. Neurocognitive Development of Motivated Behavior: Dynamic Changes across Childhood and Adolescence. J Neurosci 2018; 38:9433-9445. [PMID: 30381435 PMCID: PMC6209847 DOI: 10.1523/jneurosci.1674-18.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
The ability to anticipate and respond appropriately to the challenges and opportunities present in our environments is critical for adaptive behavior. Recent methodological innovations have led to substantial advances in our understanding of the neurocircuitry supporting such motivated behavior in adulthood. However, the neural circuits and cognitive processes that enable threat- and reward-motivated behavior undergo substantive changes over the course of development, and these changes are less well understood. In this article, we highlight recent research in human and animal models demonstrating how developmental changes in prefrontal-subcortical neural circuits give rise to corresponding changes in the processing of threats and rewards from infancy to adulthood. We discuss how these developmental trajectories are altered by experiential factors, such as early-life stress, and highlight the relevance of this research for understanding the developmental onset and treatment of psychiatric disorders characterized by dysregulation of motivated behavior.
Collapse
Affiliation(s)
- Dylan G Gee
- Department of Psychology, Yale University, New Haven, CT 06520,
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI 02912
| | - Carolyn M Johnson
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Heidi C Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065
| | - Vishnu P Murty
- Department of Psychology, Temple University, Philadelphia, PA 19122
| | - Wouter van den Bos
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, Netherlands, and
| | | |
Collapse
|
33
|
Developmental transitions in amygdala PKC isoforms and AMPA receptor expression associated with threat memory in infant rats. Sci Rep 2018; 8:14679. [PMID: 30279521 PMCID: PMC6168531 DOI: 10.1038/s41598-018-32762-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Although infants learn and remember, they rapidly forget, a phenomenon known as infantile amnesia. While myriad mechanisms impact this rapid forgetting, the molecular events supporting memory maintenance have yet to be explored. To explore memory mechanisms across development, we used amygdala-dependent odor-shock conditioning and focused on mechanisms important in adult memory, the AMPA receptor subunits GluA1/2 and upstream protein kinases important for trafficking AMPAR, protein kinase M zeta (PKMζ) and iota/lambda (PKCι/λ). We use odor-shock conditioning in infant rats because it is late-developing (postnatal day, PN10) and can be modulated by corticosterone during a sensitive period in early life. Our results show that memory-related molecules did not change in pups too young to learn threat (PN8) but were activated in pups old enough to learn (PN12), with increased PKMζ-PKCι/λ and GluA2 similar to that observed in adult memory, but with an uncharacteristic decrease in GluA1. This molecular signature and behavioral avoidance of the conditioned odor was recapitulated in PN8 pups injected with CORT before conditioning to precociously induce learning. Blocking learning via CORT inhibition in older pups (PN12) blocked the expression of these molecules. PN16 pups showed a more adult-like molecular cascade of increased PKMζ-PKCι/λ and GluA1–2. Finally, at all ages, zeta inhibitory peptide (ZIP) infusions into the amygdala 24 hr after conditioning blocked memory. Together, these results identify unique features of memory processes across early development: AMPAR subunits GluA1/2 and PKC isoform expression are differentially used, which may contribute to mechanisms of early life forgetting.
Collapse
|
34
|
Jia M, Travaglia A, Pollonini G, Fedele G, Alberini CM. Developmental changes in plasticity, synaptic, glia, and connectivity protein levels in rat medial prefrontal cortex. ACTA ACUST UNITED AC 2018; 25:533-543. [PMID: 30224556 PMCID: PMC6149953 DOI: 10.1101/lm.047753.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
The medial prefrontal cortex (mPFC) plays a critical role in complex brain functions including decision-making, integration of emotional, and cognitive aspects in memory processing and memory consolidation. Because relatively little is known about the molecular mechanisms underlying its development, we quantified rat mPFC basal expression levels of sets of plasticity, synaptic, glia, and connectivity proteins at different developmental ages. Specifically, we compared the mPFC of rats at postnatal day 17 (PN17), when they are still unable to express long-term contextual and spatial memories, to rat mPFC at PN24, when they have acquired the ability of long-term memory expression and finally to the mPFC of adult rats. We found that, with increased age, there are remarkable and significant decreases in markers of cell activation and significant increases in proteins that mark synaptogenesis and synapse maturation. Furthermore, we found significant changes in structural markers over the ages, suggesting that structural connectivity of the mPFC increases over time. Finally, the substantial biological difference in mPFC at different ages suggest caution in extrapolating conclusions from brain plasticity studies conducted at different developmental stages.
Collapse
Affiliation(s)
- Margaret Jia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Alessio Travaglia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Gabriella Pollonini
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Giuseppe Fedele
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, New York, New York 10003, USA
| |
Collapse
|
35
|
Lee S, Choi BR, Kim J, LaFerla FM, Park JHY, Han JS, Lee KW, Kim J. Sulforaphane Upregulates the Heat Shock Protein Co-Chaperone CHIP and Clears Amyloid-β and Tau in a Mouse Model of Alzheimer's Disease. Mol Nutr Food Res 2018; 62:e1800240. [PMID: 29714053 DOI: 10.1002/mnfr.201800240] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/17/2018] [Indexed: 11/08/2022]
Abstract
SCOPE Sulforaphane is an herbal isothiocyanate enriched in cruciferous vegetables. Here, the authors investigate whether sulforaphane modulates the production of amyloid-β (Aβ) and tau, the two main pathological factors in Alzheimer's disease (AD). METHODS AND RESULTS A triple transgenic mouse model of AD (3 × Tg-AD) is used to study the effect of sulforaphane. Oral gavage of sulforaphane reduces protein levels of monomeric and polymeric forms of Aβ as well as tau and phosphorylated tau in 3 × Tg-AD mice. However, sulforaphane treatment do not affect mRNA expression of amyloid precursor protein or tau. As previous studies show that Aβ and tau metabolism are influenced by a heat shock protein (HSP) co-chaperone, C-terminus of HSP70-interacting protein (CHIP), the authors examine whether sulforaphane can modulate CHIP. The authors find that sulforaphane treatment increase levels of CHIP and HSP70. Furthermore, observations of CHIP-deficient primary neurons derived from 3 × Tg-AD mice suggest that sulforaphane treatment increase CHIP level and clear the accumulation of Aβ and tau. Finally, sulforaphane ameliorated memory deficits in 3 × Tg-AD mice as reveal by novel object/location recognition tests and contextual fear conditioning tests. CONCLUSION These results demonstrate that sulforaphane treatment upregulates CHIP and has the potential to decrease the accumulation of Aβ and tau in patients with AD.
Collapse
Affiliation(s)
- Siyoung Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Bo-Ryoung Choi
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Jisung Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Frank M LaFerla
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, CA 92697, USA
| | - Jung Han Yoon Park
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung-Soo Han
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiyoung Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
36
|
Santarelli AJ, Khan AM, Poulos AM. Contextual fear retrieval-induced Fos expression across early development in the rat: An analysis using established nervous system nomenclature ontology. Neurobiol Learn Mem 2018; 155:42-49. [PMID: 29807127 DOI: 10.1016/j.nlm.2018.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/07/2018] [Accepted: 05/19/2018] [Indexed: 02/07/2023]
Abstract
The neural circuits underlying the acquisition, retention and retrieval of contextual fear conditioning have been well characterized in the adult animal. A growing body of work in younger rodents indicates that context-mediated fear expression may vary across development. However, it remains unclear how this expression may be defined across the full range of key developmental ages. Nor is it fully clear whether the structure of the adult context fear network generalizes to earlier ages. In this study, we compared context fear retrieval-induced behavior and neuroanatomically constrained immediate early-gene expression across infant (P19), early and late juvenile (P24 and P35), and adult (P90) male Long-Evans rats. We focused our analysis on neuroanatomically defined subregions and nuclei of the basolateral complex of the amygdala (BLA complex), dorsal and ventral portions of the hippocampus and the subregions of the medial prefrontal cortex as defined by the nomenclature of the Swanson (2004) adult rat brain atlas. Relative to controls and across all ages tested, there were greater numbers of Fos immunoreactive (Fos-ir) neurons in the posterior part of the basolateral amygdalar nuclei (BLAp) following context fear retrieval that correlated statistically with the expression of freezing. However, Fos-ir within regions having known connections with the BLA complex was differentially constrained by developmental age: early juvenile, but not adult rats exhibited an increase of context fear-dependent Fos-ir neurons in prelimbic and infralimbic areas, while adult, but not juvenile rats displayed increases in Fos-ir neurons within the ventral CA1 hippocampus. These results suggest that juvenile and adult rodents may recruit developmentally unique pathways in the acquisition and retrieval of contextual fear. This study extends prior work by providing a broader set of developmental ages and a rigorously defined neuroanatomical ontology within which the contextual fear network can be studied further.
Collapse
Affiliation(s)
- Anthony J Santarelli
- Department of Psychology, Center for Neuroscience, State University of New York, University at Albany, Albany, NY 12222, USA
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Andrew M Poulos
- Department of Psychology, Center for Neuroscience, State University of New York, University at Albany, Albany, NY 12222, USA.
| |
Collapse
|
37
|
Colon L, Odynocki N, Santarelli A, Poulos AM. Sexual differentiation of contextual fear responses. ACTA ACUST UNITED AC 2018; 25:230-240. [PMID: 29661835 PMCID: PMC5903402 DOI: 10.1101/lm.047159.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022]
Abstract
Development and sex differentiation impart an organizational influence on the neuroanatomy and behavior of mammalian species. Prior studies suggest that brain regions associated with fear motivated defensive behavior undergo a protracted and sex-dependent development. Outside of adult animals, evidence for developmental sex differences in conditioned fear is sparse. Here, we examined in male and female Long-Evans rats how developmental age and sex affect the long-term retention and generalization of Pavlovian fear responses. Experiments 1 and 2 describe under increasing levels of aversive learning (three and five trials) the long-term retrieval of cued and context fear in preadolescent (P24 and P33), periadolescent (P37), and adult (P60 and P90) rats. Experiments 3 and 4 examined contextual processing under minimal aversive learning (1 trial) procedures in infant (P19, P21), preadolescent (P24), and adult (P60) rats. Here, we found that male and female rats display a divergent developmental trajectory in the expression of context-mediated freezing, such that context fear expression in males tends to increase toward adulthood, while females displayed an opposite pattern of decreasing context fear expression toward adulthood. Longer (14 d) retention intervals produced an overall heightened context fear expression relative to shorter (1 d) retention intervals an observation consistent with fear incubation. Male, but not Female rats showed increasing generalization of context fear across development. Collectively, these findings provide an initial demonstration that sexual differentiation of contextual fear conditioning emerges prior to puberty and follows a distinct developmental trajectory toward adulthood that strikingly parallels sex differences in the etiology and epidemiology of anxiety and trauma- and stressor-related disorders.
Collapse
Affiliation(s)
- Lorianna Colon
- Department of Psychology and Center for Neuroscience, University at Albany, State University of New York, Albany, New York, USA
| | - Natalie Odynocki
- Department of Psychology and Center for Neuroscience, University at Albany, State University of New York, Albany, New York, USA
| | - Anthony Santarelli
- Department of Psychology and Center for Neuroscience, University at Albany, State University of New York, Albany, New York, USA
| | - Andrew M Poulos
- Department of Psychology and Center for Neuroscience, University at Albany, State University of New York, Albany, New York, USA
| |
Collapse
|
38
|
Boulanger-Bertolus J, Rincón-Cortés M, Sullivan RM, Mouly AM. Understanding pup affective state through ethologically significant ultrasonic vocalization frequency. Sci Rep 2017; 7:13483. [PMID: 29044126 PMCID: PMC5647438 DOI: 10.1038/s41598-017-13518-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022] Open
Abstract
Throughout life, rats emit ultrasonic vocalizations (USV) when confronted with an aversive situation. However, the conditions classically used to elicit USV vary greatly with the animal's age (isolation from the dam in infancy, versus nociceptive stimulation in adults). The present study is the first to characterize USV responses to the same aversive event throughout development. Specifically, infant, juvenile and adult rats were presented with mild foot-shocks and their USV frequency, duration, and relationship with respiration and behavior were compared. In juvenile and adult rats, a single class of USV is observed with an age-dependent main frequency and duration (30 kHz/400 ms in juveniles, 22 kHz/900 ms in adults). In contrast, infant rat USV were split into two classes with specific relationships with respiration and behavior: 40 kHz/300 ms and 66 kHz/21 ms. Next, we questioned if these infant USV were also emitted in a more naturalistic context by exposing pups to interactions with the mother treating them roughly. This treatment enhanced 40-kHz USV while leaving 66-kHz USV unchanged suggesting that the use of USV goes far beyond a signal studied in terms of amount of emission, and can inform us about some aspects of the infant's affective state.
Collapse
Affiliation(s)
- Julie Boulanger-Bertolus
- Lyon Neuroscience Research Center, INSERM U1028; CNRS UMR5292; University Lyon1, Lyon, France.
- University of Michigan, Ann Arbor, USA.
| | - Millie Rincón-Cortés
- Emotional Brain Institute, Nathan Kline Institute, Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, INSERM U1028; CNRS UMR5292; University Lyon1, Lyon, France
| |
Collapse
|
39
|
Debiec J, Sullivan RM. The neurobiology of safety and threat learning in infancy. Neurobiol Learn Mem 2017; 143:49-58. [PMID: 27826033 PMCID: PMC5418109 DOI: 10.1016/j.nlm.2016.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022]
Abstract
What an animal needs to learn to survive is altered dramatically as they change from dependence on the parent for protection to independence and reliance on self-defense. This transition occurs in most altricial animals, but our understanding of the behavioral neurobiology has mostly relied on the infant rat. The transformation from dependence to independence occurs over three weeks in pups and is accompanied by complex changes in responses to both natural and learned threats and the supporting neural circuitry. Overall, in early life, the threat system is quiescent and learning is biased towards acquiring attachment related behaviors to support attachment to the caregiver and proximity seeking. Caregiver-associated cues learned in infancy have the ability to provide a sense of safety throughout lifetime. This attachment/safety system is activated by learning involving presumably pleasurable stimuli (food, warmth) but also painful stimuli (tailpinch, moderate shock). At about the midway point to independence, pups begin to have access to the adult-like amygdala-dependent threat system and amygdala-dependent responses to natural dangers such as predator odors. However, pups have the ability to switch between the infant and adult-like system, which is controlled by maternal presence and modification of stress hormones. Specifically, if the pup is alone, it will learn fear but if with the mother it will learn attachment (10-15days of age). As pups begin to approach weaning, pups lose access to the attachment system and rely only on the amygdala-dependent threat system. However, pups learning system is complex and exhibits flexibility that enables the mother to override the control of the attachment circuit, since newborn pups may acquire threat responses from the mother expressing fear in their presence. Together, these data suggest that the development of pups' threat learning system is not only dependent upon maturation of the amygdala, but it is also exquisitely controlled by the environment. Most notably the mother can switch pup learning between attachment to threat learning in a moment's notice. This enables the mother to navigate pup's learning about the world and what is threatening and what is safe.
Collapse
Affiliation(s)
- Jacek Debiec
- Molecular & Behavioral Neuroscience Institute and Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States.
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Child and Adolescent Psychiatry, New York University Langone Medical Center, United States.
| |
Collapse
|
40
|
Pattwell SS, Bath KG. Emotional learning, stress, and development: An ever-changing landscape shaped by early-life experience. Neurobiol Learn Mem 2017; 143:36-48. [PMID: 28458034 PMCID: PMC5540880 DOI: 10.1016/j.nlm.2017.04.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
Abstract
The capacity to learn to associate cues with negative outcomes is a highly adaptive process that appears to be conserved across species. However, when the cue is no longer a valid predictor of danger, but the emotional response persists, this can result in maladaptive behaviors, and in humans contribute to debilitating emotional disorders. Over the past several decades, work in neuroscience, psychiatry, psychology, and biology have uncovered key processes underlying, and structures governing, emotional responding and learning, as well as identified disruptions in the structural and functional integrity of these brain regions in models of pathology. In this review, we highlight some of this elegant body of work as well as incorporate emerging findings from the field of developmental neurobiology to emphasize how development contributes to changes in the ability to learn and express emotional responses, and how early experiences, such as stress, shape the development and functioning of these circuits.
Collapse
Affiliation(s)
- Siobhan S Pattwell
- Department of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, United States.
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI 02912, United States
| |
Collapse
|
41
|
Cahill SP, Yu RQ, Green D, Todorova EV, Snyder JS. Early survival and delayed death of developmentally-born dentate gyrus neurons. Hippocampus 2017; 27:1155-1167. [PMID: 28686814 DOI: 10.1002/hipo.22760] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 01/20/2023]
Abstract
The storage and persistence of memories depends on plasticity in the hippocampus. Adult neurogenesis produces new neurons that mature through critical periods for plasticity and cellular survival, which determine their contributions to learning and memory. However, most granule neurons are generated prior to adulthood; the maturational timecourse of these neurons is poorly understood compared to adult-born neurons but is essential to identify how the dentate gyrus (DG), as a whole, contributes to behavior. To characterize neurons born in the early postnatal period, we labeled DG neurons born on postnatal day 6 (P6) with BrdU and quantified maturation and survival across early (1 hr to 8 weeks old) and late (2-6 months old) cell ages. We find that the dynamics of developmentally-born neuron survival is essentially the opposite of neurons born in adulthood: P6-born neurons did not go through a period of cell death during their immature stages (from 1 to 8 weeks). In contrast, 17% of P6-born neurons died after reaching maturity, between 2 and 6 months of age. Delayed death was evident from the loss of BrdU+ cells as well as pyknotic BrdU+ caspase3+ neurons within the superficial granule cell layer. Patterns of DCX, NeuN, and activity-dependent Fos expression indicate that developmentally-born neurons mature over several weeks and a sharp peak in zif268 expression at 2 weeks suggests that developmentally-born neurons mature faster than adult-born neurons (which peak at 3 weeks). Collectively, our findings are relevant for understanding how developmentally-born DG neurons contribute to memory and disorders throughout the lifespan. High levels of early survival and zif268 expression may promote learning, while also rendering neurons sensitive to insults at defined stages. Late neuronal death in young adulthood may result in the loss of hundreds of thousands of DG neurons, which could impact memory persistence and contribute to hippocampal/DG atrophy in disorders such as depression.
Collapse
Affiliation(s)
- Shaina P Cahill
- Department of Psychology & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ru Qi Yu
- Department of Psychology & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dylan Green
- Department of Psychology & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Evgenia V Todorova
- Department of Psychology & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason S Snyder
- Department of Psychology & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
42
|
Development of Odor Hedonics: Experience-Dependent Ontogeny of Circuits Supporting Maternal and Predator Odor Responses in Rats. J Neurosci 2017; 36:6634-50. [PMID: 27335397 DOI: 10.1523/jneurosci.0632-16.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/09/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED A major component of perception is hedonic valence: perceiving stimuli as pleasant or unpleasant. Here, we used early olfactory experiences that shape odor preferences and aversions to explore developmental plasticity in circuits mediating odor hedonics. We used 2-deoxyglucose autoradiographic mapping of neural activity to identify circuits differentially activated by biologically relevant preferred and avoided odors across rat development. We then further probed this system by increasing or decreasing hedonic value. Using both region of interest and functional connectivity analyses, we identified regions within primary olfactory, amygdala/hippocampal, and prefrontal cortical networks that were activated differentially by maternal and male odors. Although some activated regions remained stable across development (postnatal days 7-23), there was a developmental emergence of others that resulted in an age-dependent elaboration of hedonic-response-specific circuitry despite stable behavioral responses (approach/avoidance) to the odors across age. Hedonic responses to these biologically important odors were modified through diet suppression of the maternal odor and co-rearing with a male. This allowed assessment of hedonic circuits in isolation of the specific odor quality and/or intensity. Early experience significantly modified odor-evoked circuitry in an age-dependent manner. For example, co-rearing with a male, which induced pup attraction to male odor, reduced activity in amygdala regions normally activated by the unfamiliar avoided male odor, making this region more consistent with maternal odor. Understanding the development of odor hedonics, particularly within the context of altered early life experience, provides insight into the development of sensory processes, food preferences, and the formation of social affiliations, among other behaviors. SIGNIFICANCE STATEMENT Odor hedonic valence controls approach-avoidance behaviors, but also modulates ongoing behaviors ranging from food preferences and social affiliation with the caregiver to avoidance of predator odors. Experiences can shape hedonic valence. This study explored brain circuitry involved in odor hedonic encoding throughout development using maternal and predator odors and assessed the effects of early life experience on odor hedonic encoding by increasing/decreasing the hedonic value of these odors. Understanding the role of changing brain circuitry during development and its impact on behavioral function is critical for understanding sensory processing across development. These data converge with exciting literature on the brain's hedonic network and highlight the significant role of early life experience in shaping the neural networks of highly biologically relevant stimuli.
Collapse
|
43
|
Oxytocin in the amygdala and not the prefrontal cortex enhances fear and impairs extinction in the juvenile rat. Neurobiol Learn Mem 2017; 141:179-188. [DOI: 10.1016/j.nlm.2017.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 01/06/2023]
|
44
|
Opendak M, Gould E, Sullivan R. Early life adversity during the infant sensitive period for attachment: Programming of behavioral neurobiology of threat processing and social behavior. Dev Cogn Neurosci 2017; 25:145-159. [PMID: 28254197 PMCID: PMC5478471 DOI: 10.1016/j.dcn.2017.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 01/03/2017] [Accepted: 02/04/2017] [Indexed: 02/06/2023] Open
Abstract
Animals, including humans, require a highly coordinated and flexible system of social behavior and threat evaluation. However, trauma can disrupt this system, with the amygdala implicated as a mediator of these impairments in behavior. Recent evidence has further highlighted the context of infant trauma as a critical variable in determining its immediate and enduring consequences, with trauma experienced from an attachment figure, such as occurs in cases of caregiver-child maltreatment, as particularly detrimental. This review focuses on the unique role of caregiver presence during early-life trauma in programming deficits in social behavior and threat processing. Using data primarily from rodent models, we describe the interaction between trauma and attachment during a sensitive period in early life, which highlights the role of the caregiver's presence in engagement of attachment brain circuitry and suppressing threat processing by the amygdala. These data suggest that trauma experienced directly from an abusive caregiver and trauma experienced in the presence of caregiver cues produce similar neurobehavioral deficits, which are unique from those resulting from trauma alone. We go on to integrate this information into social experience throughout the lifespan, including consequences for complex scenarios, such as dominance hierarchy formation and maintenance.
Collapse
Affiliation(s)
- Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA.
| | - Elizabeth Gould
- Department of Psychology, Princeton University, Princeton, NJ, USA
| | - Regina Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA
| |
Collapse
|
45
|
Combined Supplementation of Choline and Docosahexaenoic Acid during Pregnancy Enhances Neurodevelopment of Fetal Hippocampus. Neurol Res Int 2017; 2017:8748706. [PMID: 28210506 PMCID: PMC5292192 DOI: 10.1155/2017/8748706] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/06/2016] [Accepted: 12/04/2016] [Indexed: 11/23/2022] Open
Abstract
Choline is an essential nutrient for humans which plays an important role in structural integrity and signaling functions. Docosahexaenoic acid (DHA) is a polyunsaturated fatty acid, highly enriched in cell membranes of the brain. Dietary intake of choline or DHA alone by pregnant mothers directly affects fetal brain development and function. But no studies show the efficacy of combined supplementation of choline and DHA on fetal neurodevelopment. The aim of the present study was to analyze fetal neurodevelopment on combined supplementation of pregnant dams with choline and DHA. Pregnant dams were divided into five groups: normal control [NC], saline control [SC], choline [C], DHA, and C + DHA. Saline, choline, and DHA were given as supplements to appropriate groups of dams. NC dams were undisturbed during entire gestation. On postnatal day (PND) 40, brains were processed for Cresyl staining. Pups from choline or DHA supplemented group showed significant (p < 0.05) increase in number of neurons in hippocampus when compared to the same in NC and SC groups. Moreover, pups from C + DHA supplemented group showed significantly higher number of neurons (p < 0.001) in hippocampus when compared to the same in NC and SC groups. Thus combined supplementation of choline and DHA during normal pregnancy enhances fetal hippocampal neurodevelopment better than supplementation of choline or DHA alone.
Collapse
|
46
|
Chang DJ, Debiec J. Neural correlates of the mother-to-infant social transmission of fear. J Neurosci Res 2017; 94:526-34. [PMID: 27091313 DOI: 10.1002/jnr.23739] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022]
Abstract
Although clinical and basic studies show that parental trauma, fear, and anxiety may be transmitted to offspring, the neurobiology of this transmission is still not well understood. We recently demonstrated in an animal model that infant rats acquire threat responses to a distinct cue when a mother expresses fear to this cue in their presence. This ability to acquire maternal fear through social learning is present at birth and, as we previously reported, depends on the pup's amygdala. However, the remaining neural mechanisms underlying social fear learning (SFL) in infancy remain elusive. Here, by using [(14) C]2-deoxyglucose autoradiography, we show that the mother-to-infant transmission of fear in preweaning rats is associated with a significant increase of activity in the subregions of the lateral septum, nucleus accumbens, bed nucleus of stria terminalis, retrosplenial cortex, paraventricular nucleus of the thalamus, mediodorsal and intralaminar thalamic nuclei, medial and the lateral preoptic nuclei of the hypothalamus, and the lateral periaqueductal gray. In contrast to studies of adult SFL demonstrating the role of the anterior cingulate cortex and possibly the insular cortex or research of infant classical fear conditioning showing the role of the posterior piriform cortex, no changes of activation in these areas were observed. Our results indicate that the pup's exposure to maternal fear activates a number of areas involved in processing threat, stress, or pain. This pattern of activation suggests a unique set of neural mechanisms underlying SFL in the developing brain.
Collapse
Affiliation(s)
- Da-Jeong Chang
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Jacek Debiec
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan.,Department of Psychiatry, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
47
|
Guskjolen A, Josselyn SA, Frankland PW. Age-dependent changes in spatial memory retention and flexibility in mice. Neurobiol Learn Mem 2016; 143:59-66. [PMID: 27988313 DOI: 10.1016/j.nlm.2016.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 11/26/2022]
Abstract
In humans, memories for events happening early in life are forgotten more rapidly than those for events later in life. This form of accelerated forgetting in infancy is also observed in non-human species, and has been most extensively characterized in rats. Here we expand the characterization of infantile forgetting to mice, a species where a broader range of genetic tools can be used to understand the neurobiological mechanisms underlying this form of forgetting. Using a hidden platform version of the water maze task, we first assessed retention in mice that ranged in age from 15 to 150days-old at the beginning of training. All groups exhibited spatial memory when tested one day after training. However, only mice that were 20days or older at the time of training could remember one month later. Second, forgetting in younger cohorts of mice was not due to weaker encoding, since when younger mice were over-trained, such that their performance exceeded that of adult mice, they still exhibited forgetting. Third, in young mice, presentation of a reminder one month following training led to memory recovery, indicating that forgetting was due to a retrieval, rather than storage, deficit. Fourth, younger mice exhibited superior reversal learning compared to older mice, raising the possibility that a by-product of infantile forgetting might be greater flexibility.
Collapse
Affiliation(s)
- Axel Guskjolen
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
48
|
Hunt PS, Burk JA, Barnet RC. Adolescent transitions in reflexive and non-reflexive behavior: Review of fear conditioning and impulse control in rodent models. Neurosci Biobehav Rev 2016; 70:33-45. [PMID: 27339692 PMCID: PMC5074887 DOI: 10.1016/j.neubiorev.2016.06.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/03/2016] [Accepted: 06/18/2016] [Indexed: 01/11/2023]
Abstract
Adolescence is a time of critical brain changes that pave the way for adult learning processes. However, the extent to which learning in adolescence is best characterized as a transitional linear progression from childhood to adulthood, or represents a period that differs from earlier and later developmental stages, remains unclear. Here we examine behavioral literature on associative fear conditioning and complex choice behavior with rodent models. Many aspects of fear conditioning are intact by adolescence and do not differ from adult patterns. Sufficient evidence, however, suggests that adolescent learning cannot be characterized simply as an immature precursor to adulthood. Across different paradigms assessing choice behavior, literature suggests that adolescent animals typically display more impulsive patterns of responding compared to adults. The extent to which the development of basic conditioning processes serves as a scaffold for later adult decision making is an additional research area that is important for theory, but also has widespread applications for numerous psychological conditions.
Collapse
|
49
|
|
50
|
Boulanger Bertolus J, Mouly AM, Sullivan RM. Ecologically relevant neurobehavioral assessment of the development of threat learning. Learn Mem 2016; 23:556-66. [PMID: 27634146 PMCID: PMC5026204 DOI: 10.1101/lm.042218.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/02/2016] [Indexed: 11/24/2022]
Abstract
As altricial infants gradually transition to adults, their proximate environment changes. In three short weeks, pups transition from a small world with the caregiver and siblings to a complex milieu rich in dangers as their environment expands. Such contrasting environments require different learning abilities and lead to distinct responses throughout development. Here, we will review some of the learned fear conditioned responses to threats in rats during their ontogeny, including behavioral and physiological measures that permit the assessment of learning and its supporting neurobiology from infancy through adulthood. In adulthood, odor-shock conditioning produces robust fear learning to the odor that depends upon the amygdala and related circuitry. Paradoxically, this conditioning in young pups fails to support fear learning and supports approach learning to the odor previously paired with shock. This approach learning is mediated by the infant attachment network that does not include the amygdala. During the age range when pups transition from the infant to the adult circuit (10-15 d old), pups have access to both networks: odor-shock conditioning in maternal presence uses the attachment circuit but the adult amygdala-dependent circuit when alone. However, throughout development (as young as 5 d old) the attachment associated learning can be overridden and amygdala-dependent fear learning supported, if the mother expresses fear in the presence of the pup. This social modulation of the fear permits the expression of defense reactions in life threatening situations informed by the caregiver but prevents the learning of the caregiver itself as a threat.
Collapse
Affiliation(s)
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, INSERM U1028; CNRS UMR5292; University Lyon1, Lyon, France
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York 10010, USA
| |
Collapse
|