1
|
Whittle S, Zhang L, Rakesh D. Environmental and neurodevelopmental contributors to youth mental illness. Neuropsychopharmacology 2024; 50:201-210. [PMID: 39030435 PMCID: PMC11526094 DOI: 10.1038/s41386-024-01926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/03/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
While a myriad of factors likely contribute to the development of mental illness in young people, the social environment (including early adverse experiences) in concert with neurodevelopmental alterations is undeniably important. A number of influential theories make predictions about how and why neurodevelopmental alterations may mediate or moderate the effects of the social environment on the emergence of mental illness. Here, we discuss current evidence supporting each of these theories. Although this area of research is rapidly growing, the body of evidence is still relatively limited. However, there exist some consistent findings, including increased striatal reactivity during positive affective processing and larger hippocampal volumes being associated with increased vulnerability or susceptibility to the effects of social environments on internalizing symptoms. Limited longitudinal work has investigated neurodevelopmental mechanisms linking the social environment with mental health. Drawing from human research and insights from animal studies, we propose an integrated mediation-moderation model and outline future research directions to advance the field.
Collapse
Affiliation(s)
- Sarah Whittle
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia.
- Orygen, Parkville, VIC, Australia.
| | - Lu Zhang
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Divyangana Rakesh
- Neuroimaging Department, Institute of Psychology, Psychiatry & Neuroscience, King's College London, London, UK
| |
Collapse
|
2
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
3
|
Sisk LM, Gee DG. Developmental neuroplasticity and adversity-related risk for psychopathology. Neuropsychopharmacology 2024; 50:316-317. [PMID: 39095522 PMCID: PMC11525816 DOI: 10.1038/s41386-024-01950-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Affiliation(s)
- Lucinda M Sisk
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Dylan G Gee
- Department of Psychology, Yale University, New Haven, CT, USA.
| |
Collapse
|
4
|
Ma YN, Zhang CC, Sun YX, Liu X, Li XX, Wang H, Wang T, Wang XD, Su YA, Li JT, Si TM. Dorsal CA1 NECTIN3 Reduction Mediates Early-Life Stress-Induced Object Recognition Memory Deficits in Adolescent Female Mice. Neurosci Bull 2024:10.1007/s12264-024-01305-z. [PMID: 39395912 DOI: 10.1007/s12264-024-01305-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/28/2024] [Indexed: 10/14/2024] Open
Abstract
Early-life stress (ES) leads to cognitive dysfunction in female adolescents, but the underlying neural mechanisms remain elusive. Recent evidence suggests that the cell adhesion molecules NECTIN1 and NECTIN3 play a role in cognition and ES-related cognitive deficits in male rodents. In this study, we aimed to investigate whether and how nectins contribute to ES-induced cognitive dysfunction in female adolescents. Applying the well-established limited bedding and nesting material paradigm, we found that ES impairs recognition memory, suppresses prefrontal NECTIN1 and hippocampal NECTIN3 expression, and upregulates corticotropin-releasing hormone (Crh) and its receptor 1 (Crhr1) mRNA levels in the hippocampus of adolescent female mice. Genetic experiments revealed that the reduction of dorsal CA1 (dCA1) NECTIN3 mediates ES-induced object recognition memory deficits, as knocking down dCA1 NECTIN3 impaired animals' performance in the novel object recognition task, while overexpression of dCA1 NECTIN3 successfully reversed the ES-induced deficits. Notably, prefrontal NECTIN1 knockdown did not result in significant cognitive impairments. Furthermore, acute systemic administration of antalarmin, a CRHR1 antagonist, upregulated hippocampal NECTIN3 levels and rescued object and spatial memory deficits in stressed mice. Our findings underscore the critical role of dCA1 NECTIN3 in mediating ES-induced object recognition memory deficits in adolescent female mice, highlighting it as a potential therapeutic target for stress-related psychiatric disorders in women.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| |
Collapse
|
5
|
Nieves GM, Rahn RM, Baskoylu SN, Liston CM. Divergent reward cue representations in prefrontal cortex underlie differences in reward motivation between adolescents and adults. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.07.565069. [PMID: 37986789 PMCID: PMC10659319 DOI: 10.1101/2023.11.07.565069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
A prevailing view on postnatal brain development is that brain regions gradually acquire adult functions as they mature. The medial prefrontal cortex (mPFC) regulates reward learning, motivation, and behavioral inhibition, and undergoes a protracted postnatal maturation. During adolescence, reward-seeking behavior is heightened compared to adulthood - a developmental difference that may be driven by a hypoactive mPFC, with decreased top-down control of impulsive reward-seeking. However, this hypothesis has been difficult to test directly, due in part to technical challenges of recording neuronal activity in vivo across this developmental period. Here, using a novel 2-photon imaging-compatible platform for recording mPFC activity during an operant reward conditioning task beginning early in life, we show that the adolescent mPFC is hyper-responsive to reward cues. Distinct populations of mPFC neurons encode reward-predictive cues across development, but representations of no-reward cues and unrewarded outcomes are relatively muted in adolescence. Chemogenetic inhibition of GABAergic neurons decreased motivation in adolescence but not in adulthood. Together, our findings indicate that reward-related activity in the adolescent mPFC does not gradually increase across development. On the contrary, adolescent mPFC neurons are hyper-responsive to reward-related stimuli and encode reward-predictive cues and outcomes through qualitatively different mechanisms relative to the adult mPFC, opening avenues to developing distinct, developmentally informed strategies for modulating reward-seeking behavior in adolescence and adulthood.
Collapse
Affiliation(s)
- Gabriela Manzano Nieves
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rachel M Rahn
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Saba N Baskoylu
- Picower Institute for Learning & Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Conor M Liston
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
6
|
Tooley UA, Latham A, Kenley JK, Alexopoulos D, Smyser TA, Nielsen AN, Gorham L, Warner BB, Shimony JS, Neil JJ, Luby JL, Barch DM, Rogers CE, Smyser CD. Prenatal environment is associated with the pace of cortical network development over the first three years of life. Nat Commun 2024; 15:7932. [PMID: 39256419 PMCID: PMC11387486 DOI: 10.1038/s41467-024-52242-4] [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: 08/14/2023] [Accepted: 08/30/2024] [Indexed: 09/12/2024] Open
Abstract
Environmental influences on brain structure and function during early development have been well-characterized, but whether early environments are associated with the pace of brain development is not clear. In pre-registered analyses, we use flexible non-linear models to test the theory that prenatal disadvantage is associated with differences in trajectories of intrinsic brain network development from birth to three years (n = 261). Prenatal disadvantage was assessed using a latent factor of socioeconomic disadvantage that included measures of mother's income-to-needs ratio, educational attainment, area deprivation index, insurance status, and nutrition. We find that prenatal disadvantage is associated with developmental increases in cortical network segregation, with neonates and toddlers with greater exposure to prenatal disadvantage showing a steeper increase in cortical network segregation with age, consistent with accelerated network development. Associations between prenatal disadvantage and cortical network segregation occur at the local scale and conform to a sensorimotor-association hierarchy of cortical organization. Disadvantage-associated differences in cortical network segregation are associated with language abilities at two years, such that lower segregation is associated with improved language abilities. These results shed light on associations between the early environment and trajectories of cortical development.
Collapse
Affiliation(s)
- Ursula A Tooley
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| | - Aidan Latham
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeanette K Kenley
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Tara A Smyser
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Ashley N Nielsen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Lisa Gorham
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Joshua S Shimony
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeffrey J Neil
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Joan L Luby
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Deanna M Barch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Christopher D Smyser
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
7
|
Lapp HE, Salazar M, Champagne FA. Postnatal rearing environment alters pup cues for caregiver-offspring interactions. Horm Behav 2024; 165:105630. [PMID: 39186858 DOI: 10.1016/j.yhbeh.2024.105630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Maternal behavior experienced in early life provides essential scaffolding to infant psychobiology with life-long effects on neurobiological and behavioral outcomes. However, infants are not passive recipients of caregiving. Evidence in rodents suggests that pups actively contribute to dam-pup interactions by soliciting maternal care with auditory, tactile, and hormonal cues. The limited bedding and nesting material (LBN) rearing manipulation induces changes in maternal care that have been attributed to maternal stress caused by the low-resource environment. The goal of the current study was to determine whether LBN also alters pup cues for maternal behavior, with implications for the mechanism of LBN-induced effects. Rat dams and pups were randomly assigned to LBN or Control rearing conditions on postnatal day (P) 0-6 and pups were fostered to the same or different condition on P6-13. LBN increased pup-directed maternal behaviors measured through 24 h monitoring using machine learning based automated analysis. LBN altered several pup cues known to affect maternal behavior including reducing pup core body temperature, reducing body weight, and altering pup vocalizations on P6 and P12. P6-13 LBN-exposed pups had elevated serum testosterone, which positively correlated with maternal licking and grooming. LBN reduced pup movement between nest attendance onset and the start of nursing, which was negatively related to dam nursing latency and contributed to longer nursing latency in LBN dams. P0-6 pup exposure to LBN also led to longer nest attendance bouts and shorter licking and grooming bouts on P7 and P9, suggesting lasting effects of LBN on pups. These data demonstrate that LBN changes pup behavioral and hormonal signals consistent with eliciting more maternal care, contributing to augmented pup-directed behaviors. This bidirectional interplay may be a critical mechanism involved in the lasting effects of early life environments.
Collapse
Affiliation(s)
- Hannah E Lapp
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX 78712, United States of America.
| | - Melissa Salazar
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX 78712, United States of America
| | - Frances A Champagne
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX 78712, United States of America
| |
Collapse
|
8
|
Sanguino-Gómez J, Krugers HJ. Early-life stress impairs acquisition and retrieval of fear memories: sex-effects, corticosterone modulation, and partial prevention by targeting glucocorticoid receptors at adolescent age. Neurobiol Stress 2024; 31:100636. [PMID: 38883213 PMCID: PMC11177066 DOI: 10.1016/j.ynstr.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/11/2024] [Accepted: 04/20/2024] [Indexed: 06/18/2024] Open
Abstract
The early postnatal period is a sensitive time window that is characterized by several neurodevelopmental processes that define neuronal architecture and function later in life. Here, we examined in young adult mice, using an auditory fear conditioning paradigm, whether stress during the early postnatal period 1) impacts fear acquisition and memory consolidation in male and female mice; 2) alters the fear responsiveness to corticosterone and 3) whether effects of early-life stress (ELS) can be prevented by treating mice with a glucocorticoid (GR) antagonist at adolescence. Male and female mice were exposed to a limited nesting and bedding model of ELS from postnatal day (PND) 2-9 and injected i.p with RU38486 (RU486) at adolescent age (PND 28-30). At two months of age, mice were trained in the fear conditioning (FC) paradigm (with and without post training administration of corticosterone - CORT) and freezing behavior during fear acquisition and contextual and auditory memory retrieval was scored. We observed that ELS impaired fear acquisition specifically in male mice and reduced both contextual and auditory memory retrieval in male and female mice. Acute post-training administration of CORT increased freezing levels during auditory memory retrieval in female mice but reduced freezing levels during the tone presentation in particular in control males. Treatment with RU486 prevented ELS-effects in acquisition in male mice and in females during auditory memory retrieval. In conclusion, this study highlights the long-lasting consequences of early-life stress on fear memory processing and further illustrates 1) the potential of a glucocorticoid antagonist intervention during adolescence to mitigate these effects and 2) the partial modulation of the auditory retrieval upon post training administration of CORT, with all these effects being sex-dependent.
Collapse
Affiliation(s)
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Miller JG, Gluckman PD, Fortier MV, Chong YS, Meaney MJ, Tan AP, Gotlib IH. Faster pace of hippocampal growth mediates the association between perinatal adversity and childhood depression. Dev Cogn Neurosci 2024; 67:101392. [PMID: 38761439 PMCID: PMC11127214 DOI: 10.1016/j.dcn.2024.101392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024] Open
Abstract
Early life adversity has been posited to influence the pace of structural neurodevelopment. Most research, however, has relied on cross-sectional data, which do not reveal whether the pace of neurodevelopmental change is accelerated or slowed following early exposures. In a birth cohort study that included neuroimaging data obtained at 4.5, 6, and 7.5 years of age (N = 784), we examined associations among a cumulative measure of perinatal adversity relative to resources, nonlinear trajectories of hippocampal and amygdala volume, and children's subsequent depressive symptoms at 8.5 years of age. Greater adversity was associated with reduced bilateral hippocampal body volume in early childhood, but also to faster growth in the right hippocampal body across childhood. Further, the association between adversity and childhood depressive symptoms was mediated by faster hippocampal body growth. These findings suggest that perinatal adversity is biologically embedded in hippocampal structure development, including an accelerated pace of change in the right hippocampal body that is implicated in children's psychopathology risk. In addition, our findings suggest that reduced hippocampal volume is not inconsistent with accelerated hippocampal change; these aspects of structural development may typically co-occur, as smaller regional volumes in early childhood were associated with faster growth across childhood.
Collapse
Affiliation(s)
- Jonas G Miller
- Department of Psychological Sciences, University of Connecticut, CT, USA.
| | - Peter D Gluckman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Marielle V Fortier
- Department of Diagnostic & Interventional Imaging, KK Women's and Children's Hospital, Singapore
| | - Yap Seng Chong
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, A⁎STAR Research Entities, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics & Gynecology, National University Health System, Singapore
| | - Michael J Meaney
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, A⁎STAR Research Entities, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Douglas Mental Health University Institute, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada; Brain - Body Initiative, A⁎STAR Research Entities, Singapore
| | - Ai Peng Tan
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, A⁎STAR Research Entities, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Brain - Body Initiative, A⁎STAR Research Entities, Singapore; Department of Diagnostic Imaging, National University Health System, Singapore
| | - Ian H Gotlib
- Department of Psychology, Stanford University, CA, USA
| |
Collapse
|
10
|
Cánepa ET, Berardino BG. Epigenetic mechanisms linking early-life adversities and mental health. Biochem J 2024; 481:615-642. [PMID: 38722301 DOI: 10.1042/bcj20230306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/15/2024]
Abstract
Early-life adversities, whether prenatal or postnatal exposure, have been linked to adverse mental health outcomes later in life increasing the risk of several psychiatric disorders. Research on its neurobiological consequences demonstrated an association between exposure to adversities and persistent alterations in the structure, function, and connectivity of the brain. Consistent evidence supports the idea that regulation of gene expression through epigenetic mechanisms are involved in embedding the impact of early-life experiences in the genome and mediate between social environments and later behavioral phenotypes. In addition, studies from rodent models and humans suggest that these experiences and the acquired risk factors can be transmitted through epigenetic mechanisms to offspring and the following generations potentially contributing to a cycle of disease or disease risk. However, one of the important aspects of epigenetic mechanisms, unlike genetic sequences that are fixed and unchangeable, is that although the epigenetic markings are long-lasting, they are nevertheless potentially reversible. In this review, we summarize our current understanding of the epigenetic mechanisms involved in the mental health consequences derived from early-life exposure to malnutrition, maltreatment and poverty, adversities with huge and pervasive impact on mental health. We also discuss the evidence about transgenerational epigenetic inheritance in mammals and experimental data suggesting that suitable social and pharmacological interventions could reverse adverse epigenetic modifications induced by early-life negative social experiences. In this regard, these studies must be accompanied by efforts to determine the causes that promote these adversities and that result in health inequity in the population.
Collapse
Affiliation(s)
- Eduardo T Cánepa
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| | - Bruno G Berardino
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| |
Collapse
|
11
|
Scarpa G, Antonoudiou P, Weiss G, Stone B, Maguire JL. Sex-dependent effects of early life stress on network and behavioral states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593547. [PMID: 38766016 PMCID: PMC11100797 DOI: 10.1101/2024.05.10.593547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Adverse childhood experiences (ACEs) are associated with numerous detriments in health, including increased vulnerability to psychiatric illnesses. Early life stress (ELS) in rodents has been shown to effectively model several of the behavioral and endocrine impacts of ACEs and has been utilized to investigate the underlying mechanisms contributing to disease. However, the precise neural mechanisms responsible for mediating the impact of ELS on vulnerability to psychiatric illnesses remain largely unknown. Methods We use behavior, immunoassay, in vivo LFP recording, histology, and patch clamp to describe the effects of ELS on stress behaviors, endocrinology, network states, protein expression, and cellular physiology in male and female mice. Results We demonstrate that a murine maternal separation (MS) ELS model causes sex-dependent alterations in behavioral and hormonal responses following an acute stressor. Local field potential (LFP) recordings in the basolateral amygdala (BLA) and frontal cortex (FC) reveal similar sex-dependent alterations at baseline, in response to acute ethological stress, and during fear memory extinction, supporting a large body of literature demonstrating that these network states contribute to stress reactivity and vulnerability to psychiatric illnesses. Sex differences were accompanied by altered physiology of BLA principal neurons in males and BLA PV interneurons in females. Conclusions Collectively, these results implicate novel, sex-dependent mechanisms through which ACEs may impact psychiatric health, involving altered cellular physiology and network states involved in emotional processing.
Collapse
|
12
|
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
|
13
|
Tabbaa M, Levitt P. Chd8 haploinsufficiency impacts rearing experience in C57BL/6 mice. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12892. [PMID: 38560770 PMCID: PMC10982810 DOI: 10.1111/gbb.12892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
Mutations in CHD8 are one of the highest genetic risk factors for autism spectrum disorder. Studies in mice that investigate underlying mechanisms have shown Chd8 haploinsufficient mice display some trait disruptions that mimic clinical phenotypes, although inconsistencies have been reported in some traits across different models on the same strain background. One source of variation across studies may be the impact of Chd8 haploinsufficiency on maternal-offspring interactions. While differences in maternal care as a function of Chd8 genotype have not been studied directly, a previous study showed that pup survival was reduced when reared by Chd8 heterozygous dams compared with wild-type (WT) dams, suggesting altered maternal care as a function of Chd8 genotype. Through systematic observation of the C57BL/6 strain, we first determined the impact of Chd8 haploinsufficiency in the offspring on WT maternal care frequencies across preweaning development. We next determined the impact of maternal Chd8 haploinsufficiency on pup care. Compared with litters with all WT offspring, WT dams exhibited less frequent maternal behaviors toward litters consisting of offspring with mixed Chd8 genotypes, particularly during postnatal week 1. Dam Chd8 haploinsufficiency decreased litter survival and increased active maternal care also during postnatal week 1. Determining the impact of Chd8 haploinsufficiency on early life experiences provides an important foundation for interpreting offspring outcomes and determining mechanisms that underlie heterogeneous phenotypes.
Collapse
Affiliation(s)
- Manal Tabbaa
- Children's Hospital Los AngelesThe Saban Research InstituteLos AngelesCaliforniaUSA
- Keck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Pat Levitt
- Children's Hospital Los AngelesThe Saban Research InstituteLos AngelesCaliforniaUSA
- Keck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
14
|
Spann MN, Alleyne K, Holland CM, Davids A, Pierre-Louis A, Bang C, Oyeneye V, Kiflom R, Shea E, Cheng B, Peterson BS, Monk C, Scheinost D. The effects of experience of discrimination and acculturation during pregnancy on the developing offspring brain. Neuropsychopharmacology 2024; 49:476-485. [PMID: 37968451 PMCID: PMC10724278 DOI: 10.1038/s41386-023-01765-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023]
Abstract
The experience of ethnic, racial, and structural inequalities is increasingly recognized as detrimental to health, and early studies suggest that its experience in pregnant mothers may affect the developing fetus. We characterized discrimination and acculturation experiences in a predominantly Hispanic sample of pregnant adolescent women and assessed their association with functional connectivity in their neonate's brain. We collected self-report measures of acculturation, discrimination, maternal distress (i.e., perceived stress, childhood trauma, and depressive symptoms), and socioeconomic status in 165 women. Then, we performed a data-driven clustering of acculturation, discrimination, perceived stress, depressive symptoms, trauma, and socioeconomic status variables during pregnancy to determine whether discrimination or acculturation clustered into distinct factors. Discrimination and acculturation styles loaded onto different factors from perceived stress, depressive symptoms, trauma, and socioeconomic status, suggesting that they were distinct from other factors in our sample. We associated these data-driven maternal phenotypes (discrimination and acculturation styles) with measures of resting-state functional MRI connectivity of the infant amygdala (n = 38). Higher maternal report of assimilation was associated with weaker connectivity between their neonate's amygdala and bilateral fusiform gyrus. Maternal experience of discrimination was associated with weaker connectivity between the amygdala and prefrontal cortex and stronger connectivity between the amygdala and fusiform of their neonate. Cautiously, the results may suggest a similarity to self-contained studies with adults, noting that the experience of discrimination and acculturation may influence amygdala circuitry across generations. Further prospective studies are essential that consider a more diverse population of minoritized individuals and with a comprehensive assessment of ethnic, racial, and structural factors.
Collapse
Affiliation(s)
- Marisa N Spann
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| | - Kiarra Alleyne
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Cristin M Holland
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Antonette Davids
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Arline Pierre-Louis
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Claire Bang
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | - Eileen Shea
- New York State Psychiatric Institute, New York, NY, USA
| | - Bin Cheng
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Bradley S Peterson
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Catherine Monk
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | | |
Collapse
|
15
|
Barr GA, Opendak M, Perry RE, Sarro E, Sullivan RM. Infant pain vs. pain with parental suppression: Immediate and enduring impact on brain, pain and affect. PLoS One 2023; 18:e0290871. [PMID: 37972112 PMCID: PMC10653509 DOI: 10.1371/journal.pone.0290871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/18/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND In the short term, parental presence while a human infant is in pain buffers the immediate pain responses, although emerging evidence suggests repeated social buffering of pain may have untoward long-term effects. METHODS/FINDING To explore the short- and long-term impacts of social buffering of pain, we first measured the infant rat pup's [postnatal day (PN) 8, or 12] response to mild tail shock with the mother present compared to shock alone or no shock. Shock with the mother reduced pain-related behavioral activation and USVs of pups at both ages and reduced Fos expression in the periaqueductal gray, hypothalamic paraventricular nucleus, and the amygdala at PN12 only. At PN12, shock with the mother compared to shock alone differentially regulated expression of several hundred genes related to G-protein-coupled receptors (GPCRs) and neural development, whereas PN8 pups showed a less robust and less coherent expression pattern. In a second set of experiments, pups were exposed to daily repeated Shock-mother pairings (or controls) at PN5-9 or PN10-14 (during and after pain sensitive period, respectively) and long-term outcome assessed in adults. Shock+mother pairing at PN5-9 reduced adult carrageenan-induced thermal hyperalgesia and reduced Fos expression, but PN10-14 pairings had minimal impact. The effect of infant treatment on adult affective behavior showed a complex treatment by age dependent effect. Adult social behavior was decreased following Shock+mother pairings at both PN5-9 and PN10-14, whereas shock alone had no effect. Adult fear responses to a predator odor were decreased only by PN10-14 treatment and the infant Shock alone and Shock+mother did not differ. CONCLUSIONS/SIGNIFICANCE Overall, integrating these results into our understanding of long-term programming by repeated infant pain experiences, the data suggest that pain experienced within a social context impacts infant neurobehavioral responses and initiates an altered developmental trajectory of pain and affect processing that diverges from experiencing pain alone.
Collapse
Affiliation(s)
- Gordon A. Barr
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine at the University of Pennsylvania, Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maya Opendak
- Child Study Center, Center for Early Childhood Health & Development, Child & Adolescent Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, United States of America
| | - Rosemarie E. Perry
- Child Study Center, Center for Early Childhood Health & Development, Child & Adolescent Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, United States of America
| | - Emma Sarro
- Child Study Center, Center for Early Childhood Health & Development, Child & Adolescent Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, United States of America
| | - Regina M. Sullivan
- Child Study Center, Center for Early Childhood Health & Development, Child & Adolescent Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, United States of America
| |
Collapse
|
16
|
Pardasani M, Ramakrishnan AM, Mahajan S, Kantroo M, McGowan E, Das S, Srikanth P, Pandey S, Abraham NM. Perceptual learning deficits mediated by somatostatin releasing inhibitory interneurons of olfactory bulb in an early life stress mouse model. Mol Psychiatry 2023; 28:4693-4706. [PMID: 37726451 PMCID: PMC10914616 DOI: 10.1038/s41380-023-02244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023]
Abstract
Early life adversity (ELA) causes aberrant functioning of neural circuits affecting the health of an individual. While ELA-induced behavioural disorders resulting from sensory and cognitive disabilities can be assessed clinically, the neural mechanisms need to be probed using animal models by employing multi-pronged experimental approaches. As ELA can alter sensory perception, we investigated the effect of early weaning on murine olfaction. By implementing go/no-go odour discrimination paradigm, we observed olfactory learning and memory impairments in early life stressed (ELS) male mice. As olfactory bulb (OB) circuitry plays a critical role in odour learning, we studied the plausible changes in the OB of ELS mice. Lowered c-Fos activity in the external plexiform layer and a reduction in the number of dendritic processes of somatostatin-releasing, GABAergic interneurons (SOM-INs) in the ELS mice led us to hypothesise the underlying circuit. We recorded reduced synaptic inhibitory feedback on mitral/tufted (M/T) cells, in the OB slices from ELS mice, explaining the learning deficiency caused by compromised refinement of OB output. The reduction in synaptic inhibition was nullified by the photo-activation of ChR2-expressing SOM-INs in ELS mice. The role of SOM-INs was revealed by learning-dependent refinement of Ca2+dynamics quantified by GCaMP6f signals, which was absent in ELS mice. Further, the causal role of SOM-INs involving circuitry was investigated by optogenetic modulation during the odour discrimination learning. Photo-activating these neurons rescued the ELA-induced learning deficits. Conversely, photo-inhibition caused learning deficiency in control animals, while it completely abolished the learning in ELS mice, confirming the adverse effects mediated by SOM-INs. Our results thus establish the role of specific inhibitory circuit in pre-cortical sensory area in orchestrating ELA-dependent changes.
Collapse
Affiliation(s)
- Meenakshi Pardasani
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Anantha Maharasi Ramakrishnan
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Sarang Mahajan
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Meher Kantroo
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Eleanor McGowan
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Susobhan Das
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Priyadharshini Srikanth
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Sanyukta Pandey
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
| |
Collapse
|
17
|
Nieves GM, Bravo M, Bath KG. Early life adversity ablates sex differences in active versus passive threat responding in mice. Stress 2023; 26:2244598. [PMID: 37624104 PMCID: PMC10529224 DOI: 10.1080/10253890.2023.2244598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Early life adversity (ELA) heightens the risk for anxiety disorders (which are characterized by heightened fear and avoidance behaviors), with females being twice as likely as males to develop pathology. Pavlovian fear conditioning tasks have been used to study possible mechanisms supporting endophenotypes of pathology. Identification of sex and ELA selective effects on the nature of behavioral responding in these paradigms may provide a unique window into coping strategies in response to learned fear to guide more mechanistic studies. The goals of this study were two-fold; First, to test if male and female mice employed different coping strategies in response to threat learning using different conditioning parameters (low, medium, and high intensity foot shocks). Second, to test if ELA in the form of limited bedding and nesting (LBN) altered the behavioral response of mice to conditioning. Mice received 6 tone/foot-shock pairings at one of three different foot-shock intensities (0.35 mA; 0.57 mA; 0.7 mA). Freezing, darting, and foot-shock reactivity were measured across trials. During conditioning, control-reared female mice exhibited significantly higher rates of darting behavior compared to control males at nearly all shock intensities tested. LBN rearing decreased the proportion of darting females to levels observed in males. Thus, ELA in the form of LBN significantly diminished the recruitment of active versus passive coping strategies in female mice but did not generally change male responding. Additional work will be required to understand the neural basis of these behavioral effects. Findings extending from this work have the potential to shed light on how ELA impacts trajectories of regional brain development with implications for sex-selective risk for behavioral endophenotypes associated with pathology and possibly symptom presentation.
Collapse
Affiliation(s)
- Gabriela Manzano Nieves
- Department of Psychiatry, Sackler Institute for Developmental Psychobiology, and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
| | - Marilyn Bravo
- David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, 90095
| | - Kevin G. Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute/Research Foundation for Mental Hygiene, 1051 Riverside Drive, New York, NY, 10032
- Department of Psychiatry, Columbia University Medical College, New York, NY 1003
| |
Collapse
|
18
|
Antonoudiou P, Stone B, Colmers PLW, Evans-Strong A, Walton N, Maguire J. Influence of chronic stress on network states governing valence processing: Potential relevance to the risk for psychiatric illnesses. J Neuroendocrinol 2023; 35:e13274. [PMID: 37186481 PMCID: PMC11025365 DOI: 10.1111/jne.13274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
Stress is a major risk factor for psychiatric illnesses and understanding the mechanisms through which stress disrupts behavioral states is imperative to understanding the underlying pathophysiology of mood disorders. Both chronic stress and early life stress alter valence processing, the process of assigning value to sensory inputs and experiences (positive or negative), which determines subsequent behavior and is essential for emotional processing and ultimately survival. Stress disrupts valence processing in both humans and preclinical models, favoring negative valence processing and impairing positive valence processing. Valence assignment involves neural computations performed in emotional processing hubs, including the amygdala, prefrontal cortex, and ventral hippocampus, which can be influenced by neuroendocrine mediators. Oscillations within and between these regions are critical for the neural computations necessary to perform valence processing functions. Major advances in the field have demonstrated a role for oscillatory states in valence processing under physiological conditions and emerging studies are exploring how these network states are altered under pathophysiological conditions and impacted by neuroendocrine factors. The current review highlights what is currently known regarding the impact of stress and the role of neuroendocrine mediators on network states and valence processing. Further, we propose a model in which chronic stress alters information routing through emotional processing hubs, resulting in a facilitation of negative valence processing and a suppression of positive valence processing.
Collapse
Affiliation(s)
| | - Bradly Stone
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | - Najah Walton
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jamie Maguire
- Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
20
|
Abraham M, Schmerder K, Hedtstück M, Bösing K, Mundorf A, Freund N. Maternal separation and its developmental consequences on anxiety and parvalbumin interneurons in the amygdala. J Neural Transm (Vienna) 2023; 130:1167-1175. [PMID: 37294327 PMCID: PMC10460741 DOI: 10.1007/s00702-023-02657-y] [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: 12/28/2022] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
The early postnatal period represents an exceptionally vulnerable phase for the development of neurobiological alterations, aberrant behavior, and psychiatric disorders. Altered GABAergic activity in the hippocampus and the amygdala have been identified in humans diagnosed with depression or anxiety disorders, as well as in respective animal models. Changes in GABAergic activity can be visualized by immunohistochemical staining of parvalbumin (PV) protein. Therewith, alterations in PV intensity as well as in the integrity of the perineural net surrounding PV positive (PV+) interneurons have been reported as consequences of early stress. In the current study, maternal separation (MS) was used to induce early life stress. Female and male Sprague-Dawley rats were subjected to MS over 4 h from postnatal days 2-20. Then, anxiety behavior and PV+ interneurons in the amygdala were analyzed using immunohistochemistry in adolescence or adulthood. MS induced increased anxiety behavior in the marble-burying test in adolescence as well as in the elevated plus maze in adulthood. No effect of sex was found. Concerning alterations of parvalbumin expression in the amygdala, a trend towards a lower number of parvalbumin-positive inhibitory interneurons was shown in the amygdala after MS in adolescence, with no differences in the total number of cells. The current study offers a developmental perspective, suggesting that the kind of anxiety behavior expressed by rats following MS changes over time from active to passive avoidance, indicating that effects of MS are highly dependent on developmental state. Moreover, a cell-type-specific effect of MS on the cellular composition of the amygdala is discussed. The presented study demonstrates the long-lasting consequences of early stress on behavior, offers a possible neurobiological correlate, and discusses possible mediators in the development of these alterations.
Collapse
Affiliation(s)
- Mate Abraham
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany
| | - Kirsten Schmerder
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany
| | - Malin Hedtstück
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany
| | - Kimberly Bösing
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany
| | - Annakarina Mundorf
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Universitätsstraße 150, 44780, Bochum, Germany.
| |
Collapse
|
21
|
Tooley UA, Latham A, Kenley JK, Alexopoulos D, Smyser T, Warner BB, Shimony JS, Neil JJ, Luby JL, Barch DM, Rogers CE, Smyser CD. Prenatal environment is associated with the pace of cortical network development over the first three years of life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.18.552639. [PMID: 37662189 PMCID: PMC10473645 DOI: 10.1101/2023.08.18.552639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Environmental influences on brain structure and function during early development have been well-characterized. In pre-registered analyses, we test the theory that socioeconomic status (SES) is associated with differences in trajectories of intrinsic brain network development from birth to three years (n = 261). Prenatal SES is associated with developmental increases in cortical network segregation, with neonates and toddlers from lower-SES backgrounds showing a steeper increase in cortical network segregation with age, consistent with accelerated network development. Associations between SES and cortical network segregation occur at the local scale and conform to a sensorimotor-association hierarchy of cortical organization. SES-associated differences in cortical network segregation are associated with language abilities at two years, such that lower segregation is associated with improved language abilities. These results yield key insight into the timing and directionality of associations between the early environment and trajectories of cortical development.
Collapse
Affiliation(s)
- Ursula A. Tooley
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110
| | - Aidan Latham
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110
| | - Jeanette K. Kenley
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110
| | | | - Tara Smyser
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110
| | - Barbara B. Warner
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110
| | - Joshua S. Shimony
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Jeffrey J. Neil
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Joan L. Luby
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110
| | - Deanna M. Barch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63110
| | - Cynthia E. Rogers
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110
| | - Chris D. Smyser
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
22
|
Ye Y, Mattingly MM, Sunthimer MJ, Gay JD, Rosen MJ. Early-Life Stress Impairs Perception and Neural Encoding of Rapid Signals in the Auditory Pathway. J Neurosci 2023; 43:3232-3244. [PMID: 36973014 PMCID: PMC10162457 DOI: 10.1523/jneurosci.1787-22.2023] [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: 09/19/2022] [Revised: 01/24/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
During developmental critical periods (CPs), early-life stress (ELS) induces cognitive deficits and alters neural circuitry in regions underlying learning, memory, and attention. Mechanisms underlying critical period plasticity are shared by sensory cortices and these higher neural regions, suggesting that sensory processing may also be vulnerable to ELS. In particular, the perception and auditory cortical (ACx) encoding of temporally-varying sounds both mature gradually, even into adolescence, providing an extended postnatal window of susceptibility. To examine the effects of ELS on temporal processing, we developed a model of ELS in the Mongolian gerbil, a well-established model for auditory processing. In both male and female animals, ELS induction impaired the behavioral detection of short gaps in sound, which are critical for speech perception. This was accompanied by reduced neural responses to gaps in auditory cortex, the auditory periphery, and auditory brainstem. ELS thus degrades the fidelity of sensory representations available to higher regions, and could contribute to well-known ELS-induced problems with cognition.SIGNIFICANCE STATEMENT In children and animal models, early-life stress (ELS) leads to deficits in cognition, including problems with learning, memory, and attention. Such problems could arise in part from a low-fidelity representation of sensory information available to higher-level neural regions. Here, we demonstrate that ELS degrades sensory responses to rapid variations in sound at multiple levels of the auditory pathway, and concurrently impairs perception of these rapidly-varying sounds. As these sound variations are intrinsic to speech, ELS may thus pose a challenge to communication and cognition through impaired sensory encoding.
Collapse
Affiliation(s)
- Yi Ye
- Hearing Research Group, Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
- Brain Health Research Institute, Kent State University, Kent, Ohio, 44242
| | - Michelle M Mattingly
- Hearing Research Group, Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Matthew J Sunthimer
- Hearing Research Group, Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Jennifer D Gay
- Hearing Research Group, Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
- Department of Otolaryngology, Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, 08901
| | - Merri J Rosen
- Hearing Research Group, Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
- Brain Health Research Institute, Kent State University, Kent, Ohio, 44242
| |
Collapse
|
23
|
Ferrara NC, Opendak M. Amygdala circuit transitions supporting developmentally-appropriate social behavior. Neurobiol Learn Mem 2023; 201:107762. [PMID: 37116857 PMCID: PMC10204580 DOI: 10.1016/j.nlm.2023.107762] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/30/2023] [Accepted: 04/22/2023] [Indexed: 04/30/2023]
Abstract
Social behaviors dynamically change throughout the lifespan alongside the maturation of neural circuits. The basolateral region of the amygdala (BLA), in particular, undergoes substantial maturational changes from birth throughout adolescence that are characterized by changes in excitation, inhibition, and dopaminergic modulation. In this review, we detail the trajectory through which BLA circuits mature and are influenced by dopaminergic systems to guide transitions in social behavior in infancy and adolescence using data from rodents. In early life, social behavior is oriented towards approaching the attachment figure, with minimal BLA involvement. Around weaning age, dopaminergic innervation of the BLA introduces avoidance of novel peers into rat pups' behavioral repertoire. In adolescence, social behavior transitions towards peer-peer interactions with a high incidence of social play-related behaviors. This transition coincides with an increasing role of the BLA in the regulation of social behavior. Adolescent BLA maturation can be characterized by an increasing integration and function of local inhibitory GABAergic circuits and their engagement by the medial prefrontal cortex (mPFC). Manipulation of these transitions using viral circuit dissection techniques and early adversity paradigms reveals the sensitivity of this system and its role in producing age-appropriate social behavior.
Collapse
Affiliation(s)
- Nicole C Ferrara
- Discipline of Physiology and Biophysics, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Maya Opendak
- Kennedy Krieger Institute, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA.
| |
Collapse
|
24
|
Warhaftig G, Almeida D, Turecki G. Early life adversity across different cell- types in the brain. Neurosci Biobehav Rev 2023; 148:105113. [PMID: 36863603 DOI: 10.1016/j.neubiorev.2023.105113] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Early life adversity (ELA)- which includes physical, psychological, emotional, and sexual abuse is one of the most common predictors to diverse psychopathologies later in adulthood. As ELA has a lasting impact on the brain at a developmental stage, recent findings from the field highlighted the specific contributions of different cell types to ELA and their association with long lasting consequences. In this review we will gather recent findings describing morphological, transcriptional and epigenetic alterations within neurons, glia and perineuronal nets and their associated cellular subpopulation. The findings reviewed and summarized here highlight important mechanisms underlying ELA and point to therapeutic approaches for ELA and related psychopathologies later in life.
Collapse
Affiliation(s)
- Gal Warhaftig
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal QC H3A 1A1, Canada.
| |
Collapse
|
25
|
Points of divergence on a bumpy road: early development of brain and immune threat processing systems following postnatal adversity. Mol Psychiatry 2023; 28:269-283. [PMID: 35705633 DOI: 10.1038/s41380-022-01658-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 01/11/2023]
Abstract
Lifelong indices of maladaptive behavior or illness often stem from early physiological aberrations during periods of dynamic development. This is especially true when dysfunction is attributable to early life adversity (ELA), when the environment itself is unsuitable to support development of healthy behavior. Exposure to ELA is strongly associated with atypical sensitivity and responsivity to potential threats-a characteristic that could be adaptive in situations where early adversity prepares individuals for lifelong danger, but which often manifests in difficulties with emotion regulation and social relationships. By synthesizing findings from animal research, this review will consider threat sensitivity through the lenses of associated corticolimbic brain circuitry and immune mechanisms, both of which are immature early in life to maximize adaptation for protection against environmental challenges to an individual's well-being. The forces that drive differential development of corticolimbic circuits include caretaking stimuli, physiological and psychological stressors, and sex, which influences developmental trajectories. These same forces direct developmental processes of the immune system, which bidirectionally communicates with sensory systems and emotion regulation circuits within the brain. Inflammatory signals offer a further force influencing the timing and nature of corticolimbic plasticity, while also regulating sensitivity to future threats from the environment (i.e., injury or pathogens). The early development of these systems programs threat sensitivity through juvenility and adolescence, carving paths for probable function throughout adulthood. To strategize prevention or management of maladaptive threat sensitivity in ELA-exposed populations, it is necessary to fully understand these early points of divergence.
Collapse
|
26
|
Pardo GVE, Alfaro Saca EE, Becerra Flores CT, Delgado Casós WF, Pacheco-Otalora LF. Limited bedding nesting paradigm alters maternal behavior and pup's early developmental milestones but did not induce anxiety- or depressive-like behavior in two different inbred mice. Dev Psychobiol 2023; 65:e22357. [PMID: 36567650 DOI: 10.1002/dev.22357] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
Animal models are crucial to understanding the mechanisms underlying the deleterious consequences of early-life stress. Here, we aimed to examine the effect of the limited bedding nesting (LBN) paradigm on early life development milestones and anxiety- and/or depression-like behavior in adolescent and adult mice from two inbred mice of both sexes. C57BL/6NCrl and BALB/c litters were exposed to the LBN paradigm postnatal day (PND) 2-9. Maternal behavior recording occurred on PND 3-9, and pups were weighed daily and examined to verify the eye-opening on PND 10-22. The male and female offspring underwent evaluation in the open field test, elevated plus-maze, and the forced swimming test during adolescence (PND 45-49) and adulthood (PND 75-79). We found that LBN impaired the maternal behavior patterns of both strain dams, mainly on C57BL/6NCrl strain. Also, LBN delayed the pup's eye-opening time and reduced body weight gain, impacting C57BL/6NCrl pups more. We also found that LBN decreased anxiety-related indices in adolescent and adult male but not female mice of both strains. Furthermore, LBN decreased depression-related indices only adolescent female and male BALB/c and female but not male C57BL/6NCrl mice. These findings reinforce the evidence that the LBN paradigm impairs the maternal behavior pattern and pup's early developmental milestones but does not induce anxiety- or depressive-like behavior outcomes during later life.
Collapse
Affiliation(s)
- Grace V E Pardo
- Laboratorio de Investigación en Neurociencia, Instituto Científico de Investigación, Universidad Andina del Cusco, Cuzco, Peru
| | - Eros Emanuel Alfaro Saca
- Laboratorio de Investigación en Neurociencia, Instituto Científico de Investigación, Universidad Andina del Cusco, Cuzco, Peru
| | | | - Walter Fares Delgado Casós
- Laboratorio de Investigación en Neurociencia, Instituto Científico de Investigación, Universidad Andina del Cusco, Cuzco, Peru
| | - Luis F Pacheco-Otalora
- Laboratorio de Investigación en Neurociencia, Instituto Científico de Investigación, Universidad Andina del Cusco, Cuzco, Peru
| |
Collapse
|
27
|
Banerjee T, Pati S, Tiwari P, Vaidya VA. Chronic hM3Dq-DREADD-mediated chemogenetic activation of parvalbumin-positive inhibitory interneurons in postnatal life alters anxiety and despair-like behavior in adulthood in a task- and sex-dependent manner. J Biosci 2022. [DOI: 10.1007/s12038-022-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Haikonen J, Englund J, Amarilla SP, Kharybina Z, Shintyapina A, Kegler K, Garcia MS, Atanasova T, Taira T, Hartung H, Lauri SE. Aberrant cortical projections to amygdala GABAergic neurons contribute to developmental circuit dysfunction following early life stress. iScience 2022; 26:105724. [PMID: 36582824 PMCID: PMC9792886 DOI: 10.1016/j.isci.2022.105724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Early life stress (ELS) results in enduring dysfunction of the corticolimbic circuitry, underlying emotional and social behavior. However, the neurobiological mechanisms involved remain elusive. Here, we have combined viral tracing and electrophysiological techniques to study the effects of maternal separation (MS) on frontolimbic connectivity and function in young (P14-21) rats. We report that aberrant prefrontal inputs to basolateral amygdala (BLA) GABAergic interneurons transiently increase the strength of feed-forward inhibition in the BLA, which raises LTP induction threshold in MS treated male rats. The enhanced GABAergic activity after MS exposure associates with lower functional synchronization within prefrontal-amygdala networks in vivo. Intriguingly, no differences in these parameters were detected in females, which were also resistant to MS dependent changes in anxiety-like behaviors. Impaired plasticity and synchronization during the sensitive period of circuit refinement may contribute to long-lasting functional changes in the prefrontal-amygdaloid circuitry that predispose to neuropsychiatric conditions later on in life.
Collapse
Affiliation(s)
- Joni Haikonen
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Jonas Englund
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Shyrley Paola Amarilla
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Zoia Kharybina
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Alexandra Shintyapina
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kristel Kegler
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Marta Saez Garcia
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Tsvetomira Atanasova
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Henrike Hartung
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Sari E. Lauri
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Corresponding author
| |
Collapse
|
29
|
Garvin MM, Bolton JL. Sex-specific behavioral outcomes of early-life adversity and emerging microglia-dependent mechanisms. Front Behav Neurosci 2022; 16:1013865. [PMID: 36268470 PMCID: PMC9577368 DOI: 10.3389/fnbeh.2022.1013865] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Early-life adversity (ELA) is known to alter brain circuit maturation as well as increase vulnerability to cognitive and emotional disorders. However, the importance of examining sex as a biological variable when researching the effects of ELA has not been considered until recently. This perspective discusses the sex-specific behavioral outcomes of ELA in both humans and animal models, then proposes microglia-mediated mechanisms as a potential underlying cause. Recent work in rodent models suggests that ELA provokes cognitive deficits, anhedonia, and alcohol abuse primarily in males, whereas females exhibit greater risk-taking and opioid addiction-related behaviors. In addition, emerging evidence identifies microglia as a key target of ELA. For example, we have recently shown that ELA inhibits microglial synapse engulfment and process dynamics in male mice, leading to an increase in excitatory synapse number onto corticotrophin-releasing hormone (CRH)-expressing neurons in the paraventricular nucleus of the hypothalamus (PVN) and aberrant stress responses later in life. However, ELA-induced synaptic rewiring of neural circuits differs in females during development, resulting in divergent behavioral outcomes. Thus, examining the role of microglia in the sex-specific mechanisms underlying ELA-induced neuropsychiatric disorders is an important topic for future research.
Collapse
Affiliation(s)
| | - Jessica L. Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
30
|
Scheinost D, Chang J, Lacadie C, Brennan-Wydra E, Foster R, Boxberger A, Macari S, Vernetti A, Constable RT, Ment LR, Chawarska K. Hypoconnectivity between anterior insula and amygdala associates with future vulnerabilities in social development in a neurodiverse sample of neonates. Sci Rep 2022; 12:16230. [PMID: 36171268 PMCID: PMC9517994 DOI: 10.1038/s41598-022-20617-6] [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] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
Altered resting state functional connectivity (FC) involving the anterior insula (aINS), a key node in the salience network, has been reported consistently in autism. Here we examined, for the first time, FC between the aINS and the whole brain in a sample of full-term, postmenstrual age (PMA) matched neonates (mean 44.0 weeks, SD = 1.5) who due to family history have high likelihood (HL) for developing autism (n = 12) and in controls (n = 41) without family history of autism (low likelihood, LL). Behaviors associated with autism were evaluated between 12 and 18 months (M = 17.3 months, SD = 2.5) in a subsample (25/53) of participants using the First Year Inventory (FYI). Compared to LL controls, HL neonates showed hypoconnectivity between left aINS and left amygdala. Lower connectivity between the two nodes was associated with higher FYI risk scores in the social domain (r(25) = -0.561, p = .003) and this association remained robust when maternal mental health factors were considered. Considering that a subsample of LL participants (n = 14/41) underwent brain imaging during the fetal period at PMA 31 and 34 weeks, in an exploratory analysis, we evaluated prospectively development of the LaINS-Lamy connectivity and found that the two areas strongly coactivate throughout the third trimester of pregnancy. The study identifies left lateralized anterior insula-amygdala connectivity as a potential target of further investigation into neural circuitry that enhances likelihood of future onset of social behaviors associated with autism during neonatal and potentially prenatal periods.
Collapse
Affiliation(s)
- Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, 06520, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Joseph Chang
- Department of Statistics and Data Science, Yale University, New Haven, CT, 06520, USA
| | - Cheryl Lacadie
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06510, USA
| | | | - Rachel Foster
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | | | - Suzanne Macari
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Angelina Vernetti
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - R Todd Constable
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Laura R Ment
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06510, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Katarzyna Chawarska
- Department of Statistics and Data Science, Yale University, New Haven, CT, 06520, USA.
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06510, USA.
- Yale Child Study Center, Yale School of Medicine, 300 George Street, Suite 900, New Haven, CT, 06510, USA.
| |
Collapse
|
31
|
Demaestri C, Gallo M, Mazenod E, Hong AT, Arora H, Short AK, Stern H, Baram TZ, Bath KG. Resource scarcity but not maternal separation provokes unpredictable maternal care sequences in mice and both upregulate Crh-associated gene expression in the amygdala. Neurobiol Stress 2022; 20:100484. [PMID: 36120094 PMCID: PMC9475315 DOI: 10.1016/j.ynstr.2022.100484] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/26/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for the development of pathology, including anxiety disorders. Neurodevelopmental and behavioral outcomes following ELA are multifaceted and are influenced heavily by the type of adversity experienced and sex of the individual experiencing ELA. It remains unclear what properties of ELA portend differential neurobiological risk and the basis of sex-differences for negative outcomes. Predictability of the postnatal environment has emerged as being a core feature supporting development, with the most salient signals deriving from parental care. Predictability of parental care may be a distinguishing feature of different forms of ELA, and the degree of predictability afforded by these manipulations may contribute to the diversity of outcomes observed across models. Further, questions remain as to whether differing levels of predictability may contribute to differential effects on neurodevelopment and expression of genes associated with risk for pathology. Here, we tested the hypothesis that changes in maternal behavior in mice would be contingent on the type of ELA experienced, directly comparing predictability of care in the limited bedding and nesting (LBN) and maternal separation (MS) paradigms. We then tested whether the predictability of the ELA environment altered the expression of corticotropin-releasing hormone (Crh), a sexually-dimorphic neuropeptide that regulates threat-related learning, in the amygdala of male and female mice. The LBN manipulation reliably increased the entropy of maternal care, a measure that indicates lower predictability between sequences of dam behavior. LBN and MS rearing similarly increased the frequency of nest sorties and licking of pups but had mixed effects on other aspects of dam-, pup-, and nest-related behaviors. Increased expression of Crh-related genes was observed in pups that experienced ELA, with gene expression measures showing a significant interaction with sex and type of ELA manipulation. Specifically, MS was associated with increased expression of Crh-related genes in males, but not females, and LBN primarily increased expression of these genes in females, but not males. The present study provides evidence for predictability as a distinguishing feature of models of ELA and demonstrates robust consequences of these differing experience on sex-differences in gene expression critically associated with stress responding and sex differences in risk for pathology.
Collapse
Affiliation(s)
- Camila Demaestri
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, USA
| | - Meghan Gallo
- Doctoral Program in Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, RI, USA
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, Inc./ New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Elisa Mazenod
- Doctoral Program in Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, RI, USA
| | - Alexander T. Hong
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Hina Arora
- Department of Statistics, University of California-Irvine, Irvine, CA, USA
| | - Annabel K. Short
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Hal Stern
- Department of Statistics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, CA, USA
| | - Kevin G. Bath
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, Inc./ New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
32
|
Wu X, Ding Z, Fan T, Wang K, Li S, Zhao J, Zhu W. Childhood social isolation causes anxiety-like behaviors via the damage of blood-brain barrier in amygdala in female mice. Front Cell Dev Biol 2022; 10:943067. [PMID: 36051441 PMCID: PMC9424755 DOI: 10.3389/fcell.2022.943067] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022] Open
Abstract
Social interaction plays an essential role in species survival for socialized animals. Previous studies have shown that a lack of social interaction such as social isolation, especially in the early-life phase, increases the risk of developing mental diseases in adulthood. Chronic social stress alters blood-brain barrier (BBB) integrity and increases peripheral cytokines to infiltrate the brain, which is linked to the development of depressive-like behaviors in mice, suggesting that BBB function is crucial in environmental stimuli-driven mood disorders via increased neuroinflammation in the brain. However, the precise mechanisms of inflammation and BBB integrity underlying the behavioral profiles induced by social isolation remain poorly understood. Here we showed that chronic childhood social isolation from post-weaning for consecutive 8 weeks in female but not male C57BL/6J mice induces anxiety-like behaviors. The levels of peripheral inflammatory cytokines including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the plasma of socially isolated female mice were increased. Importantly, we found decreased expression of the endothelial cell tight junction protein Claudin-5, increased BBB breakdown and microglial activation in the amygdala of isolated but not group-housed female mice. Moreover, the neuronal activity in the amygdala was increased as evidenced by c-fos positive cells, and the levels of IL-1β in the amygdala, a critical brain region for regulating social processing and interaction, were also higher in female mice exposed to social isolation. Finally, down-regulation of Claudin-5 induced anxiety-like behaviors in group-housed females and overexpression of Claudin-5 with adeno-associated virus in the amygdala to restore BBB integrity decreased subsequent anxiety-like behaviors. Together, these findings suggest that chronic childhood social isolation impaired BBB permeability and caused neuroinflammation in the amygdala by recruiting peripheral cytokines into the brain and activating microglia, consequently triggering the development of anxiety-like behaviors in female mice.
Collapse
Affiliation(s)
- Xiao Wu
- School of Basic Medical Sciences, Peking University, Beijing, China
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Zengbo Ding
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Tengteng Fan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ke Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Suxia Li
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Jing Zhao
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Jing Zhao, ; Weili Zhu,
| | - Weili Zhu
- School of Basic Medical Sciences, Peking University, Beijing, China
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
- *Correspondence: Jing Zhao, ; Weili Zhu,
| |
Collapse
|
33
|
Early life adversity shapes neural circuit function during sensitive postnatal developmental periods. Transl Psychiatry 2022; 12:306. [PMID: 35915071 PMCID: PMC9343623 DOI: 10.1038/s41398-022-02092-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for mental illness, but the neurobiological mechanisms by which ELA increases the risk for future psychopathology are still poorly understood. Brain development is particularly malleable during prenatal and early postnatal life, when complex neural circuits are being formed and refined through an interplay of excitatory and inhibitory neural input, synaptogenesis, synaptic pruning, myelination, and neurogenesis. Adversity that influences these processes during sensitive periods of development can thus have long-lasting and pervasive effects on neural circuit maturation. In this review, we will discuss clinical and preclinical evidence for the impact of ELA on neural circuit formation with a focus on the early postnatal period, and how long-lasting impairments in these circuits can affect future behavior. We provide converging evidence from human and animal studies on how ELA alters the functional development of brain regions, neural circuits, and neurotransmitter systems that are crucial for cognition and affective behavior, including the hippocampus, the hypothalamus-pituitary-adrenal (HPA) axis, neural networks of fear responses and cognition, and the serotonin (5-HT) system. We also discuss how gene-by-environment (GxE) interactions can determine individual differences in susceptibility and resilience to ELA, as well as molecular pathways by which ELA regulates neural circuit development, for which we emphasize epigenetic mechanisms. Understanding the molecular and neurobiological mechanisms underlying ELA effects on brain function and psychopathology during early postnatal sensitive periods may have great potential to advance strategies to better treat or prevent psychiatric disorders that have their origin early in life.
Collapse
|
34
|
Jeanneteau F, Coutellier L. The glucocorticoid footprint on the memory engram. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25:100378. [PMID: 38486965 PMCID: PMC10938917 DOI: 10.1016/j.coemr.2022.100378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
The complexity of the classical inverted U-shaped relationship between cortisol levels and responses transposable to stress reactivity has led to an incomplete understanding of the mechanisms enabling healthy and toxic effects of stress on brain and behavior. A clearer, more detailed, picture of those relationships can be obtained by integrating cortisol effects on large-scale brain networks, in particular, by focusing on neural network configurations from the perspective of inhibition and excitation. A unifying view of Semon and Hebb's theories of cellular memory links the biophysical and metabolic changes in neuronal ensembles to the strengthening of collective synapses. In that sense, the neuronal capacity to record, store, and retrieve information directly relates to the adaptive capacity of its connectivity and metabolic reserves. Here, we use task-activated cell ensembles or simply engram cells as an example to demonstrate that the adaptive behavioral responses to stress result from collective synapse strength within and across networks of interneurons and excitatory ones.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Laurence Coutellier
- Departments of Psychology and Neuroscience, Ohio State University, Columbus, USA
| |
Collapse
|
35
|
Effects of early life adversities upon memory processes and cognition in rodent models. Neuroscience 2022; 497:282-307. [PMID: 35525496 DOI: 10.1016/j.neuroscience.2022.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/14/2023]
Abstract
Exposure to stressors in early postnatal life induces long-lasting modifications in brainfunction.Thisplasticity,an essential characteristic of the brain that enables adaptation to the environment, may also induce impairments in some psychophysiological functions, including learning and memory. Early life stress (ELS) has long-term effects on thehypothalamic-pituitary-adrenal axisresponse to stressors, and has been reported to lead toneuroinflammation,altered levelsof neurotrophic factors, modifications inneurogenesis andsynaptic plasticity,with changes in neurotransmitter systems and network functioning. In this review, we focus on early postnatal stress in animal models and their effects on learning and memory.Many studies have reported ELS-induced impairments in different types of memories, including spatial memory, fear memory, recognition (both for objects and social) memory, working memory and reversal learning. Studies are not always in agreement, however, no effects, or sometimes facilitation, being reported, depending on the nature and intensity of the early intervention, as well as the age when the outcome was evaluated and the sex of the animals. When considering processes occurring after consolidation, related with memory maintenance or modification, there are a very reduced number of reports. Future studies addressing the mechanisms underlying memory changes for ELS should shed some light on the understanding of the different effects induced by stressors of different types and intensities on cognitive functions.
Collapse
|
36
|
Impact of stress on inhibitory neuronal circuits, our tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100460. [PMID: 35734023 PMCID: PMC9207718 DOI: 10.1016/j.ynstr.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 12/03/2022] Open
Abstract
This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.
Collapse
|
37
|
Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: Evidence from animal models of early-life adversity and addiction. Eur J Neurosci 2022; 55:2170-2195. [PMID: 33825217 PMCID: PMC8494863 DOI: 10.1111/ejn.15223] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023]
Abstract
Addiction is a chronic relapsing disorder with devastating personal, societal, and economic consequences. In humans, early-life adversity (ELA) such as trauma, neglect, and resource scarcity are linked with increased risk of later-life addiction, but the brain mechanisms underlying this link are still poorly understood. Here, we focus on data from rodent models of ELA and addiction, in which causal effects of ELA on later-life responses to drugs and the neurodevelopmental mechanisms by which ELA increases vulnerability to addiction can be determined. We first summarize evidence for a link between ELA and addiction in humans, then describe how ELA is commonly modeled in rodents. Since addiction is a heterogeneous disease with many individually varying behavioral aspects that may be impacted by ELA, we next discuss common rodent assays of addiction-like behaviors. We then summarize the specific addiction-relevant behavioral phenotypes caused by ELA in male and female rodents and discuss some of the underlying changes in brain reward and stress circuits that are likely responsible. By better understanding the behavioral and neural mechanisms by which ELA promotes addiction vulnerability, we hope to facilitate development of new approaches for preventing or treating addiction in those with a history of ELA.
Collapse
Affiliation(s)
- Sophia C. Levis
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Stephen V. Mahler
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| |
Collapse
|
38
|
Chelini G, Pangrazzi L, Bozzi Y. At the Crossroad Between Resiliency and Fragility: A Neurodevelopmental Perspective on Early-Life Experiences. Front Cell Neurosci 2022; 16:863866. [PMID: 35465609 PMCID: PMC9023311 DOI: 10.3389/fncel.2022.863866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Postnatal development of the brain is characterized by sensitive windows during which, local circuitry are drastically reshaped by life experiences. These critical periods (CPs) occur at different time points for different brain functions, presenting redundant physiological changes in the underlying brain regions. Although circuits malleability during CPs provides a valuable window of opportunity for adaptive fine-tuning to the living environment, this aspect of neurodevelopment also represents a phase of increased vulnerability for the development of a variety of disorders. Consistently, accumulating epidemiological studies point to adverse childhood experience as a major risk factor for many medical conditions, especially stress- and anxiety-related conditions. Thanks to creative approaches to manipulate rodents’ rearing environment, neurobiologist have uncovered a pivotal interaction between CPs and early-life experiences, offering an interesting landscape to improve our understanding of brain disorders. In this short review, we discuss how early-life experience impacts cellular and molecular players involved in CPs of development, translating into long-lasting behavioral consequences in rodents. Bringing together findings from multiple laboratories, we delineate a unifying theory in which systemic factors dynamically target the maturation of brain functions based on adaptive needs, shifting the balance between resilience and vulnerability in response to the quality of the rearing environment.
Collapse
Affiliation(s)
- Gabriele Chelini
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
- *Correspondence: Gabriele Chelini,
| | - Luca Pangrazzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Yuri Bozzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
- Consiglio Nazionale delle Ricerche (CNR) Neuroscience Institute, Pisa, Italy
| |
Collapse
|
39
|
Birnie MT, Levis SC, Mahler SV, Baram TZ. Developmental Trajectories of Anhedonia in Preclinical Models. Curr Top Behav Neurosci 2022; 58:23-41. [PMID: 35156184 DOI: 10.1007/7854_2021_299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This chapter discusses how the complex concept of anhedonia can be operationalized and studied in preclinical models. It provides information about the development of anhedonia in the context of early-life adversity, and the power of preclinical models to tease out the diverse molecular, epigenetic, and network mechanisms that are responsible for anhedonia-like behaviors.Specifically, we first discuss the term anhedonia, reviewing the conceptual components underlying reward-related behaviors and distinguish anhedonia pertaining to deficits in motivational versus consummatory behaviors. We then describe the repertoire of experimental approaches employed to study anhedonia-like behaviors in preclinical models, and the progressive refinement over the past decade of both experimental instruments (e.g., chemogenetics, optogenetics) and conceptual constructs (salience, valence, conflict). We follow with an overview of the state of current knowledge of brain circuits, nodes, and projections that execute distinct aspects of hedonic-like behaviors, as well as neurotransmitters, modulators, and receptors involved in the generation of anhedonia-like behaviors. Finally, we discuss the special case of anhedonia that arises following early-life adversity as an eloquent example enabling the study of causality, mechanisms, and sex dependence of anhedonia.Together, this chapter highlights the power, potential, and limitations of using preclinical models to advance our understanding of the origin and mechanisms of anhedonia and to discover potential targets for its prevention and mitigation.
Collapse
Affiliation(s)
- Matthew T Birnie
- Departments of Anatomy/Neurobiology and Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Sophia C Levis
- Departments of Anatomy/Neurobiology and Neurobiology/Behavior, University of California-Irvine, Irvine, CA, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy/Neurobiology and Pediatrics, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
40
|
Roubinov D, Meaney MJ, Boyce WT. Change of pace: How developmental tempo varies to accommodate failed provision of early needs. Neurosci Biobehav Rev 2021; 131:120-134. [PMID: 34547365 PMCID: PMC8648258 DOI: 10.1016/j.neubiorev.2021.09.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/30/2021] [Accepted: 09/16/2021] [Indexed: 01/13/2023]
Abstract
The interplay of genes and environments (GxE) is a fundamental source of variation in behavioral and developmental outcomes. Although the role of developmental time (T) in the unfolding of such interactions has yet to be fully considered, GxE operates within a temporal frame of reference across multiple timescales and degrees of biological complexity. Here, we consider GxExT interactions to understand adversity-induced developmental acceleration or deceleration whereby environmental conditions hasten or hinder children's development. To date, developmental pace changes have been largely explained through a focus on the individual: for example, how adversity "wears down" aging biological systems or how adversity accelerates or decelerates maturation to optimize reproductive fitness. We broaden such theories by positing shifts in developmental pace in response to the parent-child dyad's capacity or incapacity for meeting children's early, physiological and safety needs. We describe empirical evidence and potential neurobiological mechanisms supporting this new conceptualization of developmental acceleration and deceleration. We conclude with suggestions for future research on the developmental consequences of early adverse exposures.
Collapse
Affiliation(s)
- Danielle Roubinov
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, United States.
| | - Michael J Meaney
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec, H3H 1R4, Canada; Child and Brain Development Program, CIFAR, Toronto, Ontario, M5G 1M1, Canada; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A ⁎STAR), 117609, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
| | - W Thomas Boyce
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, United States; Child and Brain Development Program, CIFAR, Toronto, Ontario, M5G 1M1, Canada; Department of Pediatrics, University of California, San Francisco, United States
| |
Collapse
|
41
|
Ellis SN, Honeycutt JA. Sex Differences in Affective Dysfunction and Alterations in Parvalbumin in Rodent Models of Early Life Adversity. Front Behav Neurosci 2021; 15:741454. [PMID: 34803622 PMCID: PMC8600234 DOI: 10.3389/fnbeh.2021.741454] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
The early life environment markedly influences brain and behavioral development, with adverse experiences associated with increased risk of anxiety and depressive phenotypes, particularly in females. Indeed, early life adversity (ELA) in humans (i.e., caregiver deprivation, maltreatment) and rodents (i.e., maternal separation, resource scarcity) is associated with sex-specific emergence of anxious and depressive behaviors. Although these disorders show clear sex differences in humans, little attention has been paid toward evaluating sex as a biological variable in models of affective dysfunction; however, recent rodent work suggests sex-specific effects. Two widely used rodent models of ELA approximate caregiver deprivation (i.e., maternal separation) and resource scarcity (i.e., limited bedding). While these approaches model aspects of ELA experienced in humans, they span different portions of the pre-weaning developmental period and may therefore differentially contribute to underlying mechanistic risk. This is borne out in the literature, where evidence suggests differences in trajectories of behavior depending on the type of ELA and/or sex; however, the neural underpinning of these differences is not well understood. Because anxiety and depression are thought to involve dysregulation in the balance of excitatory and inhibitory signaling in ELA-vulnerable brain regions (e.g., prefrontal cortex, amygdala, hippocampus), outcomes are likely driven by alterations in local and/or circuit-specific inhibitory activity. The most abundant GABAergic subtypes in the brain, accounting for approximately 40% of inhibitory neurons, contain the calcium-binding protein Parvalbumin (PV). As PV-expressing neurons have perisomatic and proximal dendritic targets on pyramidal neurons, they are well-positioned to regulate excitatory/inhibitory balance. Recent evidence suggests that PV outcomes following ELA are sex, age, and region-specific and may be influenced by the type and timing of ELA. Here, we suggest the possibility of a combined role of PV and sex hormones driving differences in behavioral outcomes associated with affective dysfunction following ELA. This review evaluates the literature across models of ELA to characterize neural (PV) and behavioral (anxiety- and depressive-like) outcomes as a function of sex and age. Additionally, we detail a putative mechanistic role of PV on ELA-related outcomes and discuss evidence suggesting hormone influences on PV expression/function which may help to explain sex differences in ELA outcomes.
Collapse
Affiliation(s)
- Seneca N Ellis
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States
| | - Jennifer A Honeycutt
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States.,Department of Psychology, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
42
|
Gee DG. Early Adversity and Development: Parsing Heterogeneity and Identifying Pathways of Risk and Resilience. Am J Psychiatry 2021; 178:998-1013. [PMID: 34734741 DOI: 10.1176/appi.ajp.2021.21090944] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adversity early in life is common and is a major risk factor for the onset of psychopathology. Delineating the neurodevelopmental pathways by which early adversity affects mental health is critical for early risk identification and targeted treatment approaches. A rapidly growing cross-species literature has facilitated advances in identifying the mechanisms linking adversity with psychopathology, specific dimensions of adversity and timing-related factors that differentially relate to outcomes, and protective factors that buffer against the effects of adversity. Yet, vast complexity and heterogeneity in early environments and neurodevelopmental trajectories contribute to the challenges of understanding risk and resilience in the context of early adversity. In this overview, the author highlights progress in four major areas-mechanisms, heterogeneity, developmental timing, and protective factors; synthesizes key challenges; and provides recommendations for future research that can facilitate progress in the field. Translation across species and ongoing refinement of conceptual models have strong potential to inform prevention and intervention strategies that can reduce the immense burden of psychopathology associated with early adversity.
Collapse
Affiliation(s)
- Dylan G Gee
- Department of Psychology, Yale University, New Haven, Conn
| |
Collapse
|
43
|
Perlman G, Tanti A, Mechawar N. Parvalbumin interneuron alterations in stress-related mood disorders: A systematic review. Neurobiol Stress 2021; 15:100380. [PMID: 34557569 PMCID: PMC8446799 DOI: 10.1016/j.ynstr.2021.100380] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/23/2022] Open
Abstract
Stress-related psychiatric disorders including depression involve complex cellular and molecular changes in the brain, and GABAergic signaling dysfunction is increasingly implicated in the etiology of mood disorders. Parvalbumin (PV)-expressing neurons are fast-spiking interneurons that, among other roles, coordinate synchronous neuronal firing. Mounting evidence suggests that the PV neuron phenotype is altered by stress and in mood disorders. In this systematic review, we assessed PV interneuron alterations in psychiatric disorders as reported in human postmortem brain studies and animal models of environmental stress. This review aims to 1) comprehensively catalog evidence of PV cell function in mood disorders (humans) and stress models of mood disorders (animals); 2) analyze the strength of evidence of PV interneuron alterations in various brain regions in humans and rodents; 3) determine whether the modulating effect of antidepressant treatment, physical exercise, and environmental enrichment on stress in animals associates with particular effects on PV function; and 4) use this information to guide future research avenues. Its principal findings, derived mainly from rodent studies, are that stress-related changes in PV cells are only reported in a minority of studies, that positive findings are region-, age-, sex-, and stress recency-dependent, and that antidepressants protect from stress-induced apparent PV cell loss. These observations do not currently translate well to humans, although the postmortem literature on the topic remains limited.
Collapse
Affiliation(s)
| | - Arnaud Tanti
- Corresponding author. McGill Group for Suicide Studies, Department of Psychiaty, McGill University, Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| | - Naguib Mechawar
- Corresponding author. McGill Group for Suicide Studies, Department of Psychiaty, McGill University, Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| |
Collapse
|
44
|
Gunnar MR, Bowen M. What was learned from studying the effects of early institutional deprivation. Pharmacol Biochem Behav 2021; 210:173272. [PMID: 34509501 PMCID: PMC8501402 DOI: 10.1016/j.pbb.2021.173272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 11/17/2022]
Abstract
The effect of experiences in infancy on human development is a central question in developmental science. Children raised in orphanage-like institutions for their first year or so of life and then adopted into well-resourced and supportive families provide a lens on the long-term effects of early deprivation and the capacity of children to recover from this type of early adversity. While it is challenging to identify cause-and-effect relations in the study of previously institutionalized individuals, finding results that are consistent with animal experimental studies and the one randomized study of removal from institutional care support the conclusion that many of the outcomes for these children were induced by early institutional deprivation. This review examines the behavioral and neural evidence for altered executive function, declarative memory, affective disorders, reward processing, reactivity to threat, risk-taking and sensation-seeking. We then provide a brief overview of the neurobiological mechanisms that may transduce early institutional experiences into effects on brain and behavior. In addition, we discuss implications for policy and practice.
Collapse
Affiliation(s)
- Megan R Gunnar
- University of Minnesota Institute of Child Development, 51 E River Rd, Minneapolis, MN 55455, USA.
| | - Maya Bowen
- University of Minnesota Institute of Child Development, 51 E River Rd, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Gee DG, Cohodes EM. Caregiving Influences on Development: A Sensitive Period for Biological Embedding of Predictability and Safety Cues. CURRENT DIRECTIONS IN PSYCHOLOGICAL SCIENCE 2021; 30:376-383. [PMID: 34675455 DOI: 10.1177/09637214211015673] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Across species, caregivers exert a powerful influence on the neural and behavioral development of offspring. Increasingly, both animal and human research has highlighted specific patterns in caregivers' behavior that may be especially important early in life, as well as neurobiological mechanisms linking early caregiving experiences with long-term affective behavior. Here we delineate evidence for an early sensitive period during infancy and toddlerhood when caregiver inputs that are predictable and associated with safety may become biologically embedded via influences on corticolimbic circuitry involved in emotion regulation. We propose that these caregiver signals prime corticolimbic circuitry to be receptive to later stage-specific caregiver influences, such as caregivers' external regulation of children's emotional reactivity. Following caregiving adversity that disrupts predictability and safety associated with caregivers during this sensitive period, accelerated maturation of corticolimbic circuitry may foreshorten the protracted period of plasticity and caregiver influence that is characteristic of humans. This work has implications for both prevention and intervention efforts for children exposed to early life adversity.
Collapse
|
46
|
Guadagno A, Belliveau C, Mechawar N, Walker CD. Effects of Early Life Stress on the Developing Basolateral Amygdala-Prefrontal Cortex Circuit: The Emerging Role of Local Inhibition and Perineuronal Nets. Front Hum Neurosci 2021; 15:669120. [PMID: 34512291 PMCID: PMC8426628 DOI: 10.3389/fnhum.2021.669120] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
The links between early life stress (ELS) and the emergence of psychopathology such as increased anxiety and depression are now well established, although the specific neurobiological and developmental mechanisms that translate ELS into poor health outcomes are still unclear. The consequences of ELS are complex because they depend on the form and severity of early stress, duration, and age of exposure as well as co-occurrence with other forms of physical or psychological trauma. The long term effects of ELS on the corticolimbic circuit underlying emotional and social behavior are particularly salient because ELS occurs during critical developmental periods in the establishment of this circuit, its local balance of inhibition:excitation and its connections with other neuronal pathways. Using examples drawn from the human and rodent literature, we review some of the consequences of ELS on the development of the corticolimbic circuit and how it might impact fear regulation in a sex- and hemispheric-dependent manner in both humans and rodents. We explore the effects of ELS on local inhibitory neurons and the formation of perineuronal nets (PNNs) that terminate critical periods of plasticity and promote the formation of stable local networks. Overall, the bulk of ELS studies report transient and/or long lasting alterations in both glutamatergic circuits and local inhibitory interneurons (INs) and their associated PNNs. Since the activity of INs plays a key role in the maturation of cortical regions and the formation of local field potentials, alterations in these INs triggered by ELS might critically participate in the development of psychiatric disorders in adulthood, including impaired fear extinction and anxiety behavior.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Claudia Belliveau
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
47
|
Abstract
Childhood socio-economic status (SES), a measure of the availability of material and social resources, is one of the strongest predictors of lifelong well-being. Here we review evidence that experiences associated with childhood SES affect not only the outcome but also the pace of brain development. We argue that higher childhood SES is associated with protracted structural brain development and a prolonged trajectory of functional network segregation, ultimately leading to more efficient cortical networks in adulthood. We hypothesize that greater exposure to chronic stress accelerates brain maturation, whereas greater access to novel positive experiences decelerates maturation. We discuss the impact of variation in the pace of brain development on plasticity and learning. We provide a generative theoretical framework to catalyse future basic science and translational research on environmental influences on brain development.
Collapse
Affiliation(s)
- Ursula A Tooley
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, USA
- Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle S Bassett
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical and Systems Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | - Allyson P Mackey
- Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Kaul D, Schwab SG, Mechawar N, Matosin N. How stress physically re-shapes the brain: Impact on brain cell shapes, numbers and connections in psychiatric disorders. Neurosci Biobehav Rev 2021; 124:193-215. [PMID: 33556389 DOI: 10.1016/j.neubiorev.2021.01.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 01/31/2021] [Indexed: 12/16/2022]
Abstract
Severe stress is among the most robust risk factors for the development of psychiatric disorders. Imaging studies indicate that life stress is integral to shaping the human brain, especially regions involved in processing the stress response. Although this is likely underpinned by changes to the cytoarchitecture of cellular networks in the brain, we are yet to clearly understand how these define a role for stress in human psychopathology. In this review, we consolidate evidence of macro-structural morphometric changes and the cellular mechanisms that likely underlie them. Focusing on stress-sensitive regions of the brain, we illustrate how stress throughout life may lead to persistent remodelling of the both neurons and glia in cellular networks and how these may lead to psychopathology. We support that greater translation of cellular alterations to human cohorts will support parsing the psychological sequalae of severe stress and improve our understanding of how stress shapes the human brain. This will remain a critical step for improving treatment interventions and prevention outcomes.
Collapse
Affiliation(s)
- Dominic Kaul
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Sibylle G Schwab
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Naguib Mechawar
- Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia; Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| |
Collapse
|
49
|
Gee DG. Caregiving influences on emotional learning and regulation: Applying a sensitive period model. Curr Opin Behav Sci 2020; 36:177-184. [PMID: 33718534 DOI: 10.1016/j.cobeha.2020.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Early caregiving experiences play a central role in shaping corticolimbic development and emotional learning and regulation. Given dynamic changes in corticolimbic maturation, the effects of caregiving experiences are likely to depend on the developmental timing of exposure. Cross-species evidence has identified timing-related differences in the effects of caregiving adversity. However, the extent to which developmental differences in associations between caregiving adversity and corticolimbic circuitry align with a sensitive period model has remained unclear. Converging evidence from studies of caregiver deprivation points to a sensitive period for caregiving influences on corticolimbic circuitry and emotional development during infancy. By contrast, differential associations between maltreatment and corticolimbic circuitry at specific ages in childhood and adolescence may reflect experience-dependent mechanisms of plasticity. Delineating sensitive periods of development and the precise experience-related mechanisms by which caregiving experiences influence corticolimbic development is essential for refining conceptual models and understanding risk and resilience following early adversity.
Collapse
Affiliation(s)
- Dylan G Gee
- Yale University, Department of Psychology, 2 Hillhouse Avenue, New Haven, CT 06511
| |
Collapse
|
50
|
Gabard-Durnam L, McLaughlin KA. Sensitive periods in human development: charting a course for the future. Curr Opin Behav Sci 2020. [DOI: 10.1016/j.cobeha.2020.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|