1
|
Kigar SL, Cuarenta A, Zuniga CL, Chang L, Auger AP, Bakshi VP. Brain, behavior, and physiological changes associated with predator stress-An animal model for trauma exposure in adult and neonatal rats. Front Mol Neurosci 2024; 17:1322273. [PMID: 38486962 PMCID: PMC10938396 DOI: 10.3389/fnmol.2024.1322273] [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: 10/16/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
The use of predators and predator odor as stressors is an important and ecologically relevant model for studying the impact of behavioral responses to threat. Here we summarize neural substrates and behavioral changes in rats resulting from predator exposure. We briefly define the impact predator exposure has on neural targets throughout development (neonatal, juvenile, and adulthood). These findings allow us to conceptualize the impact of predator exposure in the brain, which in turn may have broader implications for human disorders such as PTSD. Importantly, inclusion of sex as a biological variable yields distinct results that may indicate neural substrates impacted by predator exposure differ based on sex.
Collapse
Affiliation(s)
- Stacey L. Kigar
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Amelia Cuarenta
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Carla L. Zuniga
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Liza Chang
- College of Agricultural and Life Sciences Academic Affairs, University of Wisconsin-Madison, Madison, WI, United States
| | - Anthony P. Auger
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, United States
| | - Vaishali P. Bakshi
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
2
|
Zuluaga MJ, Agrati D, Athaíde V, Ferreira A, Uriarte N. Fear response of rat pups to a non-aversive social stimulus: Evidence for the involvement of memory processes. Dev Psychobiol 2023; 65:e22417. [PMID: 37860902 DOI: 10.1002/dev.22417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 10/21/2023]
Abstract
Learning processes in rats during early development are importantly mediated by the mother, which represents the primary source of environmental information. This study aimed to determine whether aversive early experiences can induce the expression of pups' fear responses toward a non-aversive stimulus as a consequence of a memory process. First, we determined pups' fear responses toward an anesthetized female after being exposed to this stimulus or an empty cage together with their mothers from Postnatal Day (PNDs) 1 to 4. Second, we evaluated if the administration of the protein synthesis inhibitor cycloheximide (CHX; 0.2 mg/kg, subcutaneously (sc).) disrupted the reconsolidation processes and abolished the fear response on PND 9. Only female pups previously exposed to the female intruder expressed fear responses toward an anesthetized female on PND 8. CHX administration to female pups immediately after exposure to an anesthetized female on PND 8 suppressed fear responses on PND 9, indicating that the fear expression was the result of a memory process, probably mediated by the mother. These findings demonstrated that early experiences can shape responses to social stimuli in a sex-dependent manner and emphasize the critical role of the mother in influencing fear learning in a social context.
Collapse
Affiliation(s)
- María José Zuluaga
- Biofisicoquímica, Departamento de Ciencias Biológicas, Centro Universitario Regional Litoral Norte-Sede Salto, Universidad de la República, Salto, Uruguay
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Daniella Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Vanessa Athaíde
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Annabel Ferreira
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Uriarte
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
3
|
Feng X, Wang J, Wu J, Ren X, Zhou H, Li S, Zhang J, Wang S, Wang Y, Hu Z, Hu X, Jiang T. Abnormality of anxious behaviors and functional connectivity between the amygdala and the frontal lobe in maternally deprived monkeys. Brain Behav 2023; 13:e3027. [PMID: 37464725 PMCID: PMC10498070 DOI: 10.1002/brb3.3027] [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: 07/25/2022] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE Anxious behaviors often occur in individuals who have experienced early adversity. Anxious behaviors can bring many hazards, such as social withdrawal, eating disorders, negative self-efficacy, self-injurious thoughts and behaviors, anxiety disorders, and even depression. Abnormal behavior are is closely related to changes in corresponding circuit functions in the brain. This study investigated the relationship between brain circuits and anxious behaviors in maternal-deprived rhesus monkey animal model, which mimic early adversity in human. METHODS Twenty-five rhesus monkeys (Macaca mulatta) were grouped by two different rearing conditions: 11 normal control and mother-reared (MR) monkeys and 14 maternally deprived and peer-reared (MD) monkeys. After obtaining images of the brain areas with significant differences in maternal separation and normal control macaque function, the relationship between functional junction intensity and stereotypical behaviors was determined by correlation analysis. RESULTS The correlation analysis revealed that stereotypical behaviors were negatively correlated with the coupling between the left lateral amygdala subregion and the left inferior frontal gyrus in both MD and MR macaques. CONCLUSION This study suggests that early adversity-induced anxious behaviors are associated with changes in the strength of the amygdala-prefrontal connection. The normalization of the regions involved in the functional connection might reverse the behavioral abnormality. It provides a solid foundation for effective intervention in human early adversity. SIGNIFICANCE STATEMENT This study suggests that early adversity-induced anxious behaviors are associated with changes in the strength of the amygdala-prefrontal connection. The higher the amygdala-prefrontal connection strength, the less stereotyped behaviors exhibited by monkeys experiencing early adversity. Thus, in the future, changing the strength of the amygdala-prefrontal connection may reverse the behavioral abnormalities of individuals who experience early adversity. This study provides a solid foundation for effective intervention in humans' early adversity.
Collapse
Affiliation(s)
- Xiao‐Li Feng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Department of PhysiologyFaculty of Basic Medical ScienceKunming Medical UniversityKunmingYunnanChina
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Jiao‐Jian Wang
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jing Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Xiao‐Feng Ren
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnanChina
| | - Hui Zhou
- Department of PhysiologyFaculty of Basic Medical ScienceKunming Medical UniversityKunmingYunnanChina
| | - Si‐Yu Li
- Department of PhysiologyFaculty of Basic Medical ScienceKunming Medical UniversityKunmingYunnanChina
| | - Jie Zhang
- School of Basic Medical SciencesKunming Medical UniversityKunmingYunnanChina
| | - Sheng‐Hai Wang
- School of Basic Medical SciencesKunming Medical UniversityKunmingYunnanChina
| | - Yun Wang
- National Resource Center for Non‐Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Zheng‐Fei Hu
- National Resource Center for Non‐Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Xin‐Tian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- National Resource Center for Non‐Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Center for Excellence in Brain ScienceChinese Academy of SciencesShanghaiChina
| | - Tian‐Zi Jiang
- Brainnetome Center and National Laboratory of Pattern RecognitionInstitute of AutomationChinese Academy of SciencesBeijingChina
- Research Center for Augmented IntelligenceZhejiang LaboratoryHangzhouChina
- Center for Excellence in Brain ScienceInstitute of AutomationChinese Academy of SciencesBeijingChina
| |
Collapse
|
4
|
Stevens JS, van Rooij SJ, Stenson AF, Ely TD, Powers A, Clifford A, Kim YJ, Hinrichs R, Tottenham N, Jovanovic T. Amygdala responses to threat in violence-exposed children depend on trauma context and maternal caregiving. Dev Psychopathol 2023; 35:1159-1170. [PMID: 34689856 PMCID: PMC9069569 DOI: 10.1017/s0954579421001085] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Early life adversity (ELA) has been linked with increased arousal responses to threat, including increased amygdala reactivity. Effects of ELA on brain function are well recognized, and emerging evidence suggests that caregivers may influence how environmental stressors impact children's brain function. We investigated the hypothesis that positive interaction between mother and child can buffer against ELA effects on children's neural responses to threat, and related symptoms. N = 53 mother-child pairs (children ages 8-14 years) were recruited from an urban population at high risk for violence exposure. Maternal caregiving was measured using the Parenting Questionnaire and in a cooperation challenge task. Children viewed fearful and neutral face stimuli during functional magnetic resonance imaging. Children who experienced greater violence at home showed amygdala sensitization, whereas children experiencing more school and community violence showed amygdala habituation. Sensitization was in turn linked with externalizing symptoms. However, maternal warmth was associated with a normalization of amygdala sensitization in children, and fewer externalizing behaviors prospectively up to 1 year later. Findings suggested that the effects of violence exposure on threat-related neural circuitry depend on trauma context (inside or outside the home) and that primary caregivers can increase resilience.
Collapse
Affiliation(s)
- Jennifer S. Stevens
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Sanne J.H. van Rooij
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Anais F. Stenson
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University, Detroit, MI
| | - Timothy D. Ely
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Aimee Clifford
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Ye Ji Kim
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
| | - Rebecca Hinrichs
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Nim Tottenham
- Department of Psychology, Columbia University, New York, NY
| | - Tanja Jovanovic
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University, Detroit, MI
| |
Collapse
|
5
|
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: 15] [Impact Index Per Article: 5.0] [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
|
6
|
Formation of the Looming-evoked Innate Defensive Response during Postnatal Development in Mice. Neurosci Bull 2022; 38:741-752. [PMID: 35122602 DOI: 10.1007/s12264-022-00821-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/18/2021] [Indexed: 10/19/2022] Open
Abstract
Environmental threats often trigger innate defensive responses in mammals. However, the gradual development of functional properties of these responses during the postnatal development stage remains unclear. Here, we report that looming stimulation in mice evoked flight behavior commencing at P14-16 and had fully developed by P20-24. The visual-evoked innate defensive response was not significantly altered by sensory deprivation at an early postnatal stage. Furthermore, the percentages of wide-field and horizontal cells in the superior colliculus were notably elevated at P20-24. Our findings define a developmental time window for the formation of the visual innate defense response during the early postnatal period and provide important insight into the underlying mechanism.
Collapse
|
7
|
Gomes CI, Barr GA. Local injury and systemic infection in infants alter later nociception and pain affect during early life and adulthood. Brain Behav Immun Health 2021; 9:100175. [PMID: 34589906 PMCID: PMC8474633 DOI: 10.1016/j.bbih.2020.100175] [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: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 10/25/2022] Open
Abstract
Newborns in intensive care are regularly exposed to minor painful procedures at developmental time points when noxious stimulation would be normally absent. Pain from these interventions is inconsistently treated and often exists concurrently with systemic infection, a common comorbidity of prematurity. Our understanding of the independent and combined effects of early painful experiences and infection on pain response is incomplete. The main goals of this research therefore were to understand how pain and infection experienced early in life influence future nociceptive and affective responses to painful stimuli. Rat pups were infected with E-coli on postnatal day 2 (PN2) and had left hind paw injury with carrageenan on PN3. Standard thermal tests for acute pain, formalin tests for inflammatory pain, and conditioned place aversion testing were performed at different ages to assess the nociceptive and affective components of the pain response. Early E-coli infection and early inflammatory injury with carrageenan both independently increased pain scores following hind paw reinjury with formalin on PN8, with effects persisting into adulthood in the carrageenan exposed group. When experienced concurrently, early E-coli infection and carrageenan exposure also increased conditioned aversion to pain in adults. Effect of sex was significant only in formalin testing, with males showing higher pain scores in infancy and females showing higher pain scores as adults. These findings demonstrate that infection experienced early in life can alter both the nociceptive and affective components of the pain response and that there is a cumulative effect of local and systemic pro-inflammatory processes on the aversive component of pain.
Collapse
Affiliation(s)
- Carly I Gomes
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gordon A Barr
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, 19104, USA.,Department of Psychology, University of Pennsylvania, 425 S. University Avenue, Stephen A. Levin Building, Philadelphia, PA, USA
| |
Collapse
|
8
|
Cuarenta A, Kigar SL, Henion IC, Chang L, Bakshi VP, Auger AP. Early life stress during the neonatal period alters social play and Line1 during the juvenile stage of development. Sci Rep 2021; 11:3549. [PMID: 33574362 PMCID: PMC7878767 DOI: 10.1038/s41598-021-82953-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Early life stress (ELS) has been shown to have a significant impact on typical brain development and the manifestation of psychological disorders through epigenetic modifications that alter gene expression. Line1, a retrotransposon associated with genetic diversity, has been linked with various psychological disorders that are associated with ELS. Our previous work demonstrated altered Line1 DNA copy number in the neonatal period following stressful experiences; we therefore chose to investigate whether early life stress altered Line1 retrotransposition persists into the juvenile period of development. Our study uses a neonatal predator odor exposure (POE) paradigm to model ELS in rats. We examined Line1 using qPCR to assess Line1 expression levels and DNA copy number in the male and female juvenile amygdala, hippocampus and prefrontal cortex-areas chosen for their association with affective disorders and stress. We report a sex difference in Line1 levels within the juvenile amygdala. We also find that ELS significantly increases Line1 DNA copy number within the juvenile amygdala which correlates with reduced juvenile social play levels, suggesting the possibility that Line1 may influence juvenile social development.
Collapse
Affiliation(s)
- Amelia Cuarenta
- Department of Psychology, University of Wisconsin-Madison, Madison, USA
| | - Stacey L Kigar
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, USA
| | - Ian C Henion
- Department of Psychology, University of Wisconsin-Madison, Madison, USA
| | - Liza Chang
- Department of Psychology, University of Wisconsin-Madison, Madison, USA
| | - Vaishali P Bakshi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, USA
| | - Anthony P Auger
- Department of Psychology, University of Wisconsin-Madison, Madison, USA. .,Neuroscience Training Program, University of Wisconsin-Madison, Madison, USA.
| |
Collapse
|
9
|
Nelson CA, Gabard-Durnam LJ. Early Adversity and Critical Periods: Neurodevelopmental Consequences of Violating the Expectable Environment. Trends Neurosci 2020; 43:133-143. [PMID: 32101708 PMCID: PMC8092448 DOI: 10.1016/j.tins.2020.01.002] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/30/2019] [Accepted: 01/12/2020] [Indexed: 12/12/2022]
Abstract
It is now widely recognized that children exposed to adverse life events in the first years of life are at increased risk for a variety of neural, behavioral, and psychological sequelae. As we discuss in this paper, adverse events represent a violation of the expectable environment. If such violations occur during a critical period of brain development, the detrimental effects of early adversity are likely to be long lasting. Here we discuss the various ways adversity becomes neurobiologically embedded, and how the timing of such adversity plays an important role in determining outcomes. We conclude our paper by offering recommendations for how to elucidate the neural mechanisms responsible for the behavioral sequelae and how best to model the effects of early adversity.
Collapse
Affiliation(s)
- Charles A Nelson
- Harvard Medical School, Harvard University, Boston, MA 02115, USA; Harvard Graduate School of Education, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; Laboratories of Cognitive Neuroscience, Boston Children's Hospital, Boston, MA 02215, USA.
| | - Laurel J Gabard-Durnam
- Harvard Medical School, Harvard University, Boston, MA 02115, USA; Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
10
|
Environment and early life: Decisive factors for stress-resilience and vulnerability. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 150:155-185. [DOI: 10.1016/bs.irn.2019.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Rajan KE, Soundarya S, Karen C, Shanmugapriya V, Radhakrishnan K. Presence of Mother Reduces Early-Life Social Stress: Linking the Alteration in Hypothalamic-Pituitary-Adrenal Axis and Serotonergic System. Dev Neurosci 2019; 41:212-222. [PMID: 31865338 DOI: 10.1159/000504508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/30/2019] [Indexed: 11/19/2022] Open
Abstract
In this study, we examined whether the presence of mother suppresses early-life stressful social experience (SSE)-induced anxiety-like behavior and impairment of short-term memory later in life. On postnatal day (PND)-5, mothers with pups were grouped as follows: (i) control; (ii) maternal separation (MS); (iii) pups with mother experience the presence of a stranger (M+P-ST); and (iv) maternal separated pups experience the presence of a stranger (MSP-ST). Individuals were subjected to light-dark box and spontaneous alternation from PND-29 to 32. We observed that the MSP-ST group exhibits anxiety-like behavior and impairment in short-term memory. Further, SSE significantly elevated the adrenocorticotropic hormone, corticosterone and expression of glucocorticoid receptor (GR) in MSP-ST pups. Similarly, serotonin (5-hydroxytryptamine; 5-HT), dopamine, noradrenaline and expression of serotonin transporter levels were significantly elevated in MSP-ST pups. These observations suggest that during early postnatal days, the pups may recognize strangers by the sense of smell, and the presence of mother reduces the SSE-induced stress.
Collapse
Affiliation(s)
- Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India,
| | - Suba Soundarya
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.,School of Molecular Sciences, The University of Western Australia, Perth, Washington, Australia
| | - Christopher Karen
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Vasudevan Shanmugapriya
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Karuppasamy Radhakrishnan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.,Department of Zoology, Government Arts College, Karur, India
| |
Collapse
|
12
|
Arakawa H. Sensorimotor developmental factors influencing the performance of laboratory rodents on learning and memory. Behav Brain Res 2019; 375:112140. [PMID: 31401145 PMCID: PMC6741784 DOI: 10.1016/j.bbr.2019.112140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Behavioral studies in animal models have advanced our knowledge of brain function and the neural mechanisms of human diseases. Commonly used laboratory rodents, such as mice and rats, provide a useful tool for studying the behaviors and mechanisms associated with learning and memory processes which are cooperatively regulated by multiple underlying factors, including sensory and motor performance and emotional/defense innate components. Each of these factors shows unique ontogeny and governs the sustainment of behavioral performance in learning tasks, and thus, understanding the integrative processes of behavioral development are crucial in the accurate interpretation of the functional meaning of learning and memory behaviors expressed in commonly employed behavioral test paradigms. In this review, we will summarize the major findings in the developmental processes of rodent behavior on the basis of the emergence of fundamental components for sustaining learning and memory behaviors. Briefly, most sensory modalities (except for vision) and motor abilities are functional at the juvenile stage, in which several defensive components, including active and passive defensive strategies and risk assessment behavior, emerge. Sex differences are detectable from the juvenile stage through adulthood and are considerable factors that influence behavioral tests. The test paradigms addressed in this review include associative learning (with an emphasis on fear conditioning), spatial learning, and recognition. This basic background information will aid in accurately performing behavioral studies in laboratory rodents and will therefore contribute to reducing inappropriate interpretations of behavioral data and further advance research on learning and memory in rodent models.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St. HSF2/S251, Baltimore, MD, 21201, USA.
| |
Collapse
|
13
|
Toward an animal model of borderline personality disorder. Psychopharmacology (Berl) 2019; 236:2485-2500. [PMID: 31201478 PMCID: PMC6697600 DOI: 10.1007/s00213-019-05289-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 05/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Borderline personality disorder (BPD) is a pervasive psychiatric disorder characterized by emotion dysregulation, impulsivity, impaired self-perceptions, and interpersonal relationships and currently affects 1-3% of the US population as reported by Torgersen et al. (Arch Gen Psychiatry 58:590-596, Torgersen et al. 2001), Lenzenweger et al. (Biol Psychiatry 62:553-564, Lenzenweger et al. 2007), and Tomko et al. (J Personal Disord 28:734-750, Tomko et al. 2014). One major obstacle to our understanding of the neural underpinnings of BPD is a lack of valid animal models that translate the key known features of the disorder to a system that is amenable to study. OBJECTIVE To summarize the etiology, major symptoms, and symptom triggers of BPD and then propose a blueprint for building an animal model of BPD by choosing key components of the disorder that can be implemented in rodents. RESULTS We identify the role of early life stress and subsequent mild stress in adulthood as contributing etiological factors and the potential use of altered communication between frontal cortices and the amygdala in extinction and habituation, increased impulsivity, dysregulation of the hypothalamic pituitary axis (HPA), and increased neuroinflammation as biological markers of BPD. Building upon these features of BPD, we propose a two-hit animal model that uses maternal abandonment to alter maturation of the HPA axis and mild secondary adult stress to evoke behavioral symptoms such as increased impulsivity and impaired extinction, habituation, and social interactions. CONCLUSION Through exploration of the etiology, symptom presentation, and altered neurological function, we propose an animal model of BPD. We believe that a number of existing animal paradigms that model other mental health disorders should be combined in a unique way to reflect the etiology, symptom presentation, and altered neurological function that is evident in BPD. These model, when compared with available human data, will inform research and treatment in humans for better understanding of systems from the micro-molecular level to more global physiology underlying BPD.
Collapse
|
14
|
Exposure to maternal odor enhances intake of a taste that mimicks the sensory attributes of ethanol. Learn Behav 2019; 47:302-309. [PMID: 31264146 DOI: 10.3758/s13420-019-00373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Early exposure to ethanol increases subsequent acceptance of this drug. Little attention, however, has been devoted to the interaction of the taste of the drug with other, familiar or non-familiar, odors contingent with ethanol access, particularly early in ontogeny. This study assessed the influence of exposure to maternal odor on intake and grasp responses to an artificial nipple providing a solution (a sucrose-quinine mix) that emulates the taste of alcohol, in 4-day-old rat pups. The results showed that the mother's odor enhanced intake from and seeking responses to an artificial nipple that provided the solution that mimicked the taste of alcohol (Experiment 1). This pattern of results was not evoked by the odor of an unrelated dam (Experiment 2), nor was it observed when the nipple delivered water. The main new finding of the present study is that 4-day-old rats tested in the presence of the mother (and hence exposed to her odor cues) exhibited enhanced seeking and intake of a solution that mimics the chemosensory properties of ethanol.
Collapse
|
15
|
Junod A, Opendak M, LeDoux JE, Sullivan RM. Development of Threat Expression Following Infant Maltreatment: Infant and Adult Enhancement but Adolescent Attenuation. Front Behav Neurosci 2019; 13:130. [PMID: 31293397 PMCID: PMC6603125 DOI: 10.3389/fnbeh.2019.00130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Early life maltreatment by the caregiver constitutes a major risk factor for the development of later-life psychopathologies, including fear-related pathologies. Here, we used an animal model of early life maltreatment induced by the Scarcity-Adversity Model of low bedding (LB) where the mother is given insufficient bedding for nest building while rat pups were postnatal days (PN) 8-12. To assess effects of maltreatment on the expression of threat-elicited defensive behaviors, animals underwent odor-shock threat conditioning at three developmental stages: late infancy (PN18), adolescence (PN45) or adulthood (>PN75) and tested the next day with odor only presentations (cue test). Results showed that in typically developing rats, the response to threat increases with maturation, although experience with maltreatment in early infancy produced enhanced responding to threat in infancy and adulthood, but a decrease in maltreated adolescents. To better understand the unique features of this decreased threat responding in adolescence, c-Fos expression was assessed within the amygdala and ventromedial prefrontal cortex (vmPFC) associated with the cued expression of threat learning. Fos counts across amygdala subregions were lower in LB rats compared to controls, while enhanced c-Fos expression was observed in the vmPFC prelimbic cortex (PL). Correlational analysis between freezing behavior and Fos revealed freezing levels were correlated with CeA in controls, although more global correlations were detected in LB-reared rats, including the BA, LA, and CeA. Functional connectivity analysis between brain regions showed that LB reared rats exhibited more diffuse interconnectivity across amygdala subnuclei, compared the more heterogeneous patterns observed in controls. In addition, functional connectivity between the IL and LA switched from positive to negative in abused adolescents. Overall, these results suggest that in adolescence, the unique developmental decrease in fear expression following trauma is associated with distinct changes in regional function and long-range connectivity, reminiscent of pathological brain function. These results suggest that early life maltreatment from the caregiver perturbs the developmental trajectory of threat-elicited behavior. Indeed, it is possible that this form of trauma, where the infant's safety signal or "safe haven" (the caregiver) is actually the source of the threat, produces distinct outcomes across development.
Collapse
Affiliation(s)
- Anouchka Junod
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Joseph E. LeDoux
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| |
Collapse
|
16
|
Cowan CSM, Stylianakis AA, Richardson R. Early-life stress, microbiota, and brain development: probiotics reverse the effects of maternal separation on neural circuits underpinning fear expression and extinction in infant rats. Dev Cogn Neurosci 2019; 37:100627. [PMID: 30981894 PMCID: PMC6969299 DOI: 10.1016/j.dcn.2019.100627] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/17/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022] Open
Abstract
Early-life stress has pervasive, typically detrimental, effects on physical and mental health across the lifespan. In rats, maternal-separation stress results in premature expression of an adult-like profile of fear regulation that predisposes stressed rats to persistent fear, one of the hallmarks of clinical anxiety. Probiotic treatment attenuates the effects of maternal separation on fear regulation. However, the neural pathways underlying these behavioral changes are unknown. Here, we examined the neural correlates of stress-induced alterations in fear behavior and their reversal by probiotic treatment. Male Sprague-Dawley rats were exposed to either standard rearing conditions or maternal-separation stress (postnatal days [P] 2–14). Some maternally-separated (MS) animals were also exposed to probiotics (Lactobacillus rhamnosus and L. helveticus) via the maternal drinking water during the period of stress. Using immunohistochemistry, we demonstrated that stressed rat pups prematurely exhibit adult-like engagement of the medial prefrontal cortex during fear regulation, an effect that can be prevented using a probiotic treatment. The present results add to the cross-species evidence that early adversity hastens maturation in emotion-related brain circuits. Importantly, our results also demonstrate that the precocious neural maturation in stressed infants is prevented by a non-invasive probiotic treatment.
Collapse
Affiliation(s)
- Caitlin S M Cowan
- School of Psychology, The University of New South Wales, Sydney, Australia.
| | | | - Rick Richardson
- School of Psychology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
17
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
18
|
Karen C, Rajan KE. Social Behaviour and Epigenetic Status in Adolescent and Adult Rats: The Contribution of Early-Life Stressful Social Experience. Cell Mol Neurobiol 2019; 39:371-385. [PMID: 30710320 PMCID: PMC11479603 DOI: 10.1007/s10571-019-00655-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/24/2019] [Indexed: 01/14/2023]
Abstract
Early-life experiences have been linked to individual's epigenetic status and social behaviour. Therefore, the present study aims to test whether the presence of mother suppress the early-life stressful social experience (SSE)-induced effect on social behaviour of adolescent and adult rats, and associated epigenetic changes. To test this, experimental groups [maternally separated pups (MSP)/pups with their mother (M+P)] were allowed to experience the presence of a stranger (ST), and then their social behaviour was compared with the maternal separated (MS) and control (Con) group. We observed that MS, MSP-ST group showed less social interaction with the unknown conspecifics than known conspecifics compared to other groups. Subsequently, we found that SSE elevated the level of DNA methyltransferases (Dnmt3a), ten-eleven translocation (Tet3), methyl-CpG-binding protein-2 (MeCP2) and Repressor Element-1 Silencing Transcription Factor (REST) in amygdala of adolescent and adult MS, MSP-ST groups compared to other groups. As expected, SSE altered the histone (H3) lysine (K14/K9) acetylation (ac) and H3K4/K9 methylation (me2/me3). SSE decreased the level of H3K14ac and H3K9ac in adolescents and then increased in adults. Interestingly, H3K4me2/me3 levels were elevated in adolescent and adults. Whereas H3K9me2/me3 shows contrasting pattern in adolescent, but H3K9me2/me3 levels were increased in adults. In addition, the expression of brain-derived neurotrophic factor (BDNF) was reduced in MS, MSP-ST groups' adolescent and adult rats. Observed correlation between epigenetic changes and social behaviour possibly contributed by early-life SSE in the absence of mother, but mother's presence suppresses the effect of early-life SSE.
Collapse
Affiliation(s)
- Christopher Karen
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India.
| |
Collapse
|
19
|
Age and sex differences in the innate defensive behaviors of C57BL/6 mice exhibited in a fear conditioning paradigm and upon exposure to a predatory odor. Physiol Behav 2019; 204:264-274. [PMID: 30840847 DOI: 10.1016/j.physbeh.2019.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/23/2019] [Indexed: 12/29/2022]
Abstract
Defensive responses of mice include an array of specific behaviors that involve adaptive components based on the assessment of the threat of confrontation. The freezing response is represented by a motionless moment without any specific posture or behavioral sequence, and it is widely used in the fear conditioning paradigms and other relevant defensive situations. However, freezing measurements include fragmental components of several defensive behaviors that are exhibited during the session, such as behavioral inhibition, crouching, and a moment of risk assessment behavior. From an ethological view, behavioral analyses of C57BL/6 mice of both sexes and three different ages (postnatal days (P) 25, 35, and 65) revealed a rich variety of defensive behaviors during a fear conditioning session and in response to predatory odor exposure as a nonconditioned behavior. P-25 and 35 mice exhibited more behavioral inhibition than P-65 adult mice, and P-65 mice exhibited a crouching posture more often than younger mice. This age difference was more pronounced in males. The stretch-attend posture (SAP) increased with age, except in P-25 males, which exhibited robust SAP in response to a conditioned cue; this response indicates that P-25 males are defensive in a situation-nonmatching manner. Situation-dependent defense strategies were revealed in P-35 and 65 mice: Fear conditioning paradigm induced more robust defensive responses than predatory odor exposure, to which mice primarily exhibited SAP. A sex-based difference was revealed in adult mice. Males tended to show more passive defensive responses, such as crouching and withdrawal, and females exhibited more active responses, such as SAP. These age- and sex-based differences may stem from the ethological demands and illustrate adolescent ontogenetic processes of defense behavior.
Collapse
|
20
|
Mitra R. Neuronal Plasticity in the Amygdala Following Predator Stress Exposure. Front Behav Neurosci 2019; 13:25. [PMID: 30842731 PMCID: PMC6391327 DOI: 10.3389/fnbeh.2019.00025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/30/2019] [Indexed: 11/13/2022] Open
Abstract
Predation causes robust long-term stress-related effects on prey individuals even if they do not get consumed by the predator. Here I review the role of basolateral amygdala (BLA) neurons in the mediation of non-consumptive effects of predation. This brain region is critical for the generation and maintenance of fear response across many phylogenetic groups. The exposure to cues of predator presence activates neurons within the BLA. Hormones secreted during stressful episodes cause long-lasting structural changes in BLA neurons, causing facilitation of endocrine response during subsequent exposure to stressful episodes like later predator exposure. Some studies also suggest that BLA is involved in creating anticipatory defensive behavior in response to the expectation of change in the environment.
Collapse
Affiliation(s)
- Rupshi Mitra
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
21
|
Sleep Impact on Perception, Memory, and Emotion in Adults and the Effects of Early-Life Experience. HANDBOOK OF SLEEP RESEARCH 2019. [DOI: 10.1016/b978-0-12-813743-7.00039-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
22
|
Abuaish S, Spinieli RL, McGowan PO. Perinatal high fat diet induces early activation of endocrine stress responsivity and anxiety-like behavior in neonates. Psychoneuroendocrinology 2018; 98:11-21. [PMID: 30086533 DOI: 10.1016/j.psyneuen.2018.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/01/2018] [Accepted: 08/01/2018] [Indexed: 12/21/2022]
Abstract
The maternal environment has a profound effect on the development of offspring, including responses to stress mediated by the hypothalamic-pituitary-adrenal (HPA) axis. In rodents, perinatal high fat diet (HFD) has been shown to program the HPA axis in a manner that persists throughout adulthood, however the effects of perinatal HFD on stress-related behaviors and physiology in neonates are limited. The first two weeks of life in rodents are known as the stress hyporesponsive period, during which animals do not respond to stressors that are otherwise known to elicit behavioral and physiological responses in mature animals. As neonates emerge from the hyporesponsive period, the maturing neural systems mediating the HPA axis leads to the suppression of ultrasonic vocalizations (USVs) and movement in the presence of threatening stimuli, such as male adult rat odor. In this study, we investigated the effects of perinatal HFD exposure, spanning the maternal pregestation, gestation and lactation period, on stress-related behaviors and physiology in neonatal rat offspring throughout the stress hyporesponsive period. During the stress hyporesponsive period, postnatal day (PND) 7, HFD pups had higher corticosterone levels in response to the presence of male odor, produced fewer USVs, and had an increase in basal corticotropin releasing hormone (Crh) transcript levels in the paraventricular nucleus of the hypothalamus. As pup emerged from the stress hyporesponsive period, PND 13, HFD offspring exhibited higher adrenocorticotropic hormone (ACTH) levels in response to male odor, increased anxiety-like behaviors as shown by increased USVs and immobility, and lower glucocorticoid receptor (Nr3c1) transcript abundance in the ventral hippocampus. These results indicate an alteration in the typical physiological and behavioral responses to stress during the hyporesponsive period of the HPA axis as a function of perinatal HFD exposure, which involves changes in the regulation of key genes mediating the HPA axis.
Collapse
Affiliation(s)
- Sameera Abuaish
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Department of Cell and Systems Biology, University of Toronto, Scarborough Campus, 1265 Military Trail, Toronto, ON, Canada
| | - Richard L Spinieli
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Department of Cell and Systems Biology, University of Toronto, Scarborough Campus, 1265 Military Trail, Toronto, ON, Canada; Psychobiology Graduate Program, School of Philosophy, Science and Literature of Ribeirão Preto of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Patrick O McGowan
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Department of Cell and Systems Biology, University of Toronto, Scarborough Campus, 1265 Military Trail, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
23
|
|
24
|
Gee DG, Bath KG, Johnson CM, Meyer HC, Murty VP, van den Bos W, Hartley CA. Neurocognitive Development of Motivated Behavior: Dynamic Changes across Childhood and Adolescence. J Neurosci 2018; 38:9433-9445. [PMID: 30381435 PMCID: PMC6209847 DOI: 10.1523/jneurosci.1674-18.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
The ability to anticipate and respond appropriately to the challenges and opportunities present in our environments is critical for adaptive behavior. Recent methodological innovations have led to substantial advances in our understanding of the neurocircuitry supporting such motivated behavior in adulthood. However, the neural circuits and cognitive processes that enable threat- and reward-motivated behavior undergo substantive changes over the course of development, and these changes are less well understood. In this article, we highlight recent research in human and animal models demonstrating how developmental changes in prefrontal-subcortical neural circuits give rise to corresponding changes in the processing of threats and rewards from infancy to adulthood. We discuss how these developmental trajectories are altered by experiential factors, such as early-life stress, and highlight the relevance of this research for understanding the developmental onset and treatment of psychiatric disorders characterized by dysregulation of motivated behavior.
Collapse
Affiliation(s)
- Dylan G Gee
- Department of Psychology, Yale University, New Haven, CT 06520,
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI 02912
| | - Carolyn M Johnson
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Heidi C Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065
| | - Vishnu P Murty
- Department of Psychology, Temple University, Philadelphia, PA 19122
| | - Wouter van den Bos
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, Netherlands, and
| | | |
Collapse
|
25
|
Developmental transitions in amygdala PKC isoforms and AMPA receptor expression associated with threat memory in infant rats. Sci Rep 2018; 8:14679. [PMID: 30279521 PMCID: PMC6168531 DOI: 10.1038/s41598-018-32762-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Although infants learn and remember, they rapidly forget, a phenomenon known as infantile amnesia. While myriad mechanisms impact this rapid forgetting, the molecular events supporting memory maintenance have yet to be explored. To explore memory mechanisms across development, we used amygdala-dependent odor-shock conditioning and focused on mechanisms important in adult memory, the AMPA receptor subunits GluA1/2 and upstream protein kinases important for trafficking AMPAR, protein kinase M zeta (PKMζ) and iota/lambda (PKCι/λ). We use odor-shock conditioning in infant rats because it is late-developing (postnatal day, PN10) and can be modulated by corticosterone during a sensitive period in early life. Our results show that memory-related molecules did not change in pups too young to learn threat (PN8) but were activated in pups old enough to learn (PN12), with increased PKMζ-PKCι/λ and GluA2 similar to that observed in adult memory, but with an uncharacteristic decrease in GluA1. This molecular signature and behavioral avoidance of the conditioned odor was recapitulated in PN8 pups injected with CORT before conditioning to precociously induce learning. Blocking learning via CORT inhibition in older pups (PN12) blocked the expression of these molecules. PN16 pups showed a more adult-like molecular cascade of increased PKMζ-PKCι/λ and GluA1–2. Finally, at all ages, zeta inhibitory peptide (ZIP) infusions into the amygdala 24 hr after conditioning blocked memory. Together, these results identify unique features of memory processes across early development: AMPAR subunits GluA1/2 and PKC isoform expression are differentially used, which may contribute to mechanisms of early life forgetting.
Collapse
|
26
|
Bassey RB, Gondré-Lewis MC. Combined early life stressors: Prenatal nicotine and maternal deprivation interact to influence affective and drug seeking behavioral phenotypes in rats. Behav Brain Res 2018; 359:814-822. [PMID: 30055209 DOI: 10.1016/j.bbr.2018.07.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022]
Abstract
Early life stress (ELS) increases the risk for later cognitive and emotional dysfunction, and has been implicated in the etiology of multiple psychiatric disorders. We hypothesize that combined insults during gestation and infancy, critical periods of neural development, could exacerbate neuropsychiatric outcomes in later life. Thus, we investigated the effects of maternal deprivation (MD) stress alone or combined with prenatal nicotine exposure (PNE) on negative affective states, ethanol drinking, and development of mesolimbic loci that regulate depression and drug dependence. On the elevated plus maze (EPM), MD rats exhibited ∼50% increase in risk-taking behavior/decreased anxiety when compared to control, but the combined MD + PNE did not affect this specific behavior. In the open field test, however, both MD and MD + PNE groups showed 2-fold greater locomotor activity. Furthermore, whereas MD showed greater latency to fall at 40 RPM on the rotarod compared to control, the MD + PNE animals' latency to fall was significantly greater at all RPMs tested, with an approximate 15% enhancement in motor coordination overall compared to control and MD. Analyses of depressive symptomatology with the forced swim test (FST) yielded 2- and 3-fold higher immobility times in MD and MD + PNE respectively. When tested in an operant drinking paradigm to quantify the effect of treatment on 10%v/v ethanol drinking, the MD and MD + PNE groups showed heightened ethanol consumption by ∼3- and 2-fold respectively. However, the experience of PNE reduced ethanol consumption in adults relative to MD alone. To test the stressors' impact on neurons in the amygdala and ventral tegmental area (VTA), mesolimbic anatomical regions associated with mood and reward, unbiased stereological measurements were performed and revealed ∼15% increase in number and density of neurons in the amygdala for both MD and MD + PNE, and ∼13% reduction in dopaminergic-like neurons in the VTA compared to control. We report here that multiple early stressors including prenatal nicotine and MD can modulate the neuroanatomy of the amygdala and VTA. These early life stressors can interact to influence the development of depressive-like and addictive behaviors.
Collapse
Affiliation(s)
- Rosemary B Bassey
- Department of Anatomy, Howard University, College of Medicine, Washington D.C. 20059, USA; Department of Psychiatry and Behavioral Sciences, Howard University, College of Medicine, Washington D.C. 20059, USA
| | - Marjorie C Gondré-Lewis
- Department of Anatomy, Howard University, College of Medicine, Washington D.C. 20059, USA; Department of Psychiatry and Behavioral Sciences, Howard University, College of Medicine, Washington D.C. 20059, USA.
| |
Collapse
|
27
|
Niermann HCM, Figner B, Tyborowska A, Cillessen AHN, Roelofs K. Investigation of the Stability of Human Freezing-Like Responses to Social Threat From Mid to Late Adolescence. Front Behav Neurosci 2018; 12:97. [PMID: 29867396 PMCID: PMC5964744 DOI: 10.3389/fnbeh.2018.00097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/25/2018] [Indexed: 12/15/2022] Open
Abstract
Freezing behavior, a commonly observed defensive stress response, shows relatively high stability over time in animals. Given the relevance of freezing for stress-coping and human psychopathology, it is relevant to know whether freezing behavior is also stable in humans, particularly during adolescence, when most affective symptoms develop. In a prospective longitudinal study, we investigated freezing-like behavior in response to social threat in 75 adolescents at age 14, repeated 3 years later at age 17. We used a well-established method combining electrocardiography (ECG; heart rate) and posturography (body sway) in response to emotional picture-viewing of angry, happy, and neutral faces. We hypothesized that individual differences in freezing-like behavior in response to social threat—operationalized by contrasting angry vs. neutral faces—would be relatively stable over time. Our results indeed showed relative stability between ages 14 and 17 in individual differences in freezing-like behavior in heart rate (r = 0.82), as well as in combined heart rate and body sway measures (r = 0.65). These effects were not specific for the angry vs. neutral contrast; they were also visible in other emotion contrasts. Exploratory analysis in males and females separately showed stability in body sway specifically for angry vs. neutral faces only in females. Together, these results suggest moderate to strong stability in human freezing-like behavior in response to social threat from mid to late adolescence (with exception for the body sway measure in males). This relative stability was not specific for threat-induction and may reflect a general stability that is particularly strong for heart rate. The fact that this relative stability was found over a relatively long time range of 3 years is promising for studies aiming to use freezing-like behavior as a marker for internalizing symptoms in adolescent development.
Collapse
Affiliation(s)
- Hannah C M Niermann
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Bernd Figner
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Anna Tyborowska
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | | | - Karin Roelofs
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
28
|
Debiec J, Sullivan RM. The neurobiology of safety and threat learning in infancy. Neurobiol Learn Mem 2017; 143:49-58. [PMID: 27826033 PMCID: PMC5418109 DOI: 10.1016/j.nlm.2016.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022]
Abstract
What an animal needs to learn to survive is altered dramatically as they change from dependence on the parent for protection to independence and reliance on self-defense. This transition occurs in most altricial animals, but our understanding of the behavioral neurobiology has mostly relied on the infant rat. The transformation from dependence to independence occurs over three weeks in pups and is accompanied by complex changes in responses to both natural and learned threats and the supporting neural circuitry. Overall, in early life, the threat system is quiescent and learning is biased towards acquiring attachment related behaviors to support attachment to the caregiver and proximity seeking. Caregiver-associated cues learned in infancy have the ability to provide a sense of safety throughout lifetime. This attachment/safety system is activated by learning involving presumably pleasurable stimuli (food, warmth) but also painful stimuli (tailpinch, moderate shock). At about the midway point to independence, pups begin to have access to the adult-like amygdala-dependent threat system and amygdala-dependent responses to natural dangers such as predator odors. However, pups have the ability to switch between the infant and adult-like system, which is controlled by maternal presence and modification of stress hormones. Specifically, if the pup is alone, it will learn fear but if with the mother it will learn attachment (10-15days of age). As pups begin to approach weaning, pups lose access to the attachment system and rely only on the amygdala-dependent threat system. However, pups learning system is complex and exhibits flexibility that enables the mother to override the control of the attachment circuit, since newborn pups may acquire threat responses from the mother expressing fear in their presence. Together, these data suggest that the development of pups' threat learning system is not only dependent upon maturation of the amygdala, but it is also exquisitely controlled by the environment. Most notably the mother can switch pup learning between attachment to threat learning in a moment's notice. This enables the mother to navigate pup's learning about the world and what is threatening and what is safe.
Collapse
Affiliation(s)
- Jacek Debiec
- Molecular & Behavioral Neuroscience Institute and Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States.
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Child and Adolescent Psychiatry, New York University Langone Medical Center, United States.
| |
Collapse
|
29
|
Development of Odor Hedonics: Experience-Dependent Ontogeny of Circuits Supporting Maternal and Predator Odor Responses in Rats. J Neurosci 2017; 36:6634-50. [PMID: 27335397 DOI: 10.1523/jneurosci.0632-16.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/09/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED A major component of perception is hedonic valence: perceiving stimuli as pleasant or unpleasant. Here, we used early olfactory experiences that shape odor preferences and aversions to explore developmental plasticity in circuits mediating odor hedonics. We used 2-deoxyglucose autoradiographic mapping of neural activity to identify circuits differentially activated by biologically relevant preferred and avoided odors across rat development. We then further probed this system by increasing or decreasing hedonic value. Using both region of interest and functional connectivity analyses, we identified regions within primary olfactory, amygdala/hippocampal, and prefrontal cortical networks that were activated differentially by maternal and male odors. Although some activated regions remained stable across development (postnatal days 7-23), there was a developmental emergence of others that resulted in an age-dependent elaboration of hedonic-response-specific circuitry despite stable behavioral responses (approach/avoidance) to the odors across age. Hedonic responses to these biologically important odors were modified through diet suppression of the maternal odor and co-rearing with a male. This allowed assessment of hedonic circuits in isolation of the specific odor quality and/or intensity. Early experience significantly modified odor-evoked circuitry in an age-dependent manner. For example, co-rearing with a male, which induced pup attraction to male odor, reduced activity in amygdala regions normally activated by the unfamiliar avoided male odor, making this region more consistent with maternal odor. Understanding the development of odor hedonics, particularly within the context of altered early life experience, provides insight into the development of sensory processes, food preferences, and the formation of social affiliations, among other behaviors. SIGNIFICANCE STATEMENT Odor hedonic valence controls approach-avoidance behaviors, but also modulates ongoing behaviors ranging from food preferences and social affiliation with the caregiver to avoidance of predator odors. Experiences can shape hedonic valence. This study explored brain circuitry involved in odor hedonic encoding throughout development using maternal and predator odors and assessed the effects of early life experience on odor hedonic encoding by increasing/decreasing the hedonic value of these odors. Understanding the role of changing brain circuitry during development and its impact on behavioral function is critical for understanding sensory processing across development. These data converge with exciting literature on the brain's hedonic network and highlight the significant role of early life experience in shaping the neural networks of highly biologically relevant stimuli.
Collapse
|
30
|
Early life stress and later peer distress on depressive behavior in adolescent female rats: Effects of a novel intervention on GABA and D2 receptors. Behav Brain Res 2017; 330:37-45. [PMID: 28499915 DOI: 10.1016/j.bbr.2017.04.053] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/19/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
Abstract
Early life adversity (ELA) increases the risk of depression during adolescence that may result from a decline in parvalbumin (PVB) secondary to increased neuroinflammation. In this study, we investigated depressive-like behavior following exposure to two different types of stressors that are relevant for their developmental period: 1) chronic ELA (maternal separation; MS) and 2) an acute emotional stressor during adolescence (witnessing their peers receive multiple shocks; WIT), and their interaction. We also determined whether reducing inflammation by cyclooxygenase-2 (COX-2) inhibition would prevent the onset of depressive-like behavior. Female Sprague-Dawley rat pups underwent MS for four-hours/day or received typical care (CON) between postnatal days (P) 2 and P20. A COX-2 inhibitor (COX-2I) or vehicle was administered every other day between P30 and P38. Subjects were tested for learned helplessness to assess depressive-like behavior at P40 (adolescence). MS females demonstrated increased escape latency and decreased PVB in the prefrontal cortex (PFC) and dorsal raphe that were attenuated by COX-2I intervention. Helplessness was also associated with an increase in D2 receptors in the accumbens. In contrast, WIT elevated escape latency in CON, but reduced latency in MS females. Furthermore, COX-2I intervention decreased escape latency in both CON and MS after WIT. WIT reduced PVB levels in the basolateral amygdala and increased PFC levels to CON levels. Our data suggest that decreased PVB in the PFC is important for the expression of depressive-like behavior and suggest that COX-2I intervention may provide a novel prevention for depression.
Collapse
|
31
|
Silvers JA, Goff B, Gabard-Durnam LJ, Gee DG, Fareri DS, Caldera C, Tottenham N. Vigilance, the Amygdala, and Anxiety in Youths with a History of Institutional Care. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 2:493-501. [PMID: 28993819 DOI: 10.1016/j.bpsc.2017.03.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Early adversity is commonly associated with alterations of amygdala circuitry and increased anxiety. While many theoretical and clinical accounts of early adversity suggest that it increases vigilance to threatening stimuli, the present study tested whether heightened anxiety and amygdala reactivity associated with early adversity enhanced goal-directed attention for threatening stimuli. Showing this association would provide support that these adversity-induced alterations are developmental adaptations of the individual. METHODS 34 children and adolescents who experienced early adversity in the form of previous institutionalization (PI) (26 female, mean age=13.49 years) and a comparison group of 33 children and adolescents who were reared by their biological parents since birth (16 female, mean age=13.40 years) underwent fMRI scanning while completing a visual search task that involved quickly locating a negative (fearful face) or positive target (happy face) in an array of neutral distractor stimuli (neutral faces). RESULTS Across both groups, individual differences in vigilant behavior were positively associated with amygdala responses for negative versus positive stimuli. However, a moderation analysis revealed that the degree to which amygdala responses were greater for negative versus positive stimuli was associated with greater anxiety symptomology for PI youth, but not comparison youth. CONCLUSIONS Together, these findings suggest that institutional care strengthens linkages between amygdala reactivity and anxiety, perhaps serving to enhance goal-directed attention. The findings are discussed as both adaptations as well as risk to the individual.
Collapse
Affiliation(s)
- Jennifer A Silvers
- Department of Psychology, University of California-Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095
| | - Bonnie Goff
- Department of Psychology, University of California-Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095
| | - Laurel J Gabard-Durnam
- Department of Psychology, Columbia University, 1190 Amsterdam Avenue, New York, NY 10027
| | - Dylan G Gee
- Department of Psychology, Yale University, Box 208205, New Haven, CT 06520-8205
| | - Dominic S Fareri
- Gordon F. Derner Institute of Advanced Psychological Studies, Adelphi University, 1 South Avenue, Garden City, NY 11530
| | - Christina Caldera
- Department of Psychology, University of California-Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095
| | - Nim Tottenham
- Department of Psychology, Columbia University, 1190 Amsterdam Avenue, New York, NY 10027
| |
Collapse
|
32
|
Santiago A, Aoki C, Sullivan RM. From attachment to independence: Stress hormone control of ecologically relevant emergence of infants' responses to threat. Curr Opin Behav Sci 2017; 14:78-85. [PMID: 28239630 DOI: 10.1016/j.cobeha.2016.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Young infant rat pups learn to approach cues associated with pain rather than learning amygdala-dependent fear. This approach response is considered caregiver-seeking and ecologically relevant within the context of attachment. With maturation, increases in the stress hormone corticosterone permit amygdala-dependent fear, which is crucial for survival during independent living. During the developmental transition from attachment to fear learning, maternal presence suppresses corticosterone elevation to block amygdala-dependent fear learning and re-engage the attachment circuitry. Early life trauma disrupts this developmental sequence by triggering a precocious increase of corticosterone, which permits amygdala-dependent threat responses. In this review, we explore the importance of the stress hormone corticosterone in infants' transition from complete dependence on the caregiver to independence, with consideration for environmental influences on threat response ontogeny and mechanistic importance of social buffering of the stress response.
Collapse
Affiliation(s)
- Adrienne Santiago
- Emotional Brain Institute, Nathan Kline Institute, New York University Langone Medical Center, New York, NY 10003; Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY 10003; Center for Neural Science, New York University, New York, NY 10003
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, NY 10003
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, New York University Langone Medical Center, New York, NY 10003; Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY 10003; Center for Neural Science, New York University, New York, NY 10003
| |
Collapse
|
33
|
Opendak M, Gould E, Sullivan R. Early life adversity during the infant sensitive period for attachment: Programming of behavioral neurobiology of threat processing and social behavior. Dev Cogn Neurosci 2017; 25:145-159. [PMID: 28254197 PMCID: PMC5478471 DOI: 10.1016/j.dcn.2017.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 01/03/2017] [Accepted: 02/04/2017] [Indexed: 02/06/2023] Open
Abstract
Animals, including humans, require a highly coordinated and flexible system of social behavior and threat evaluation. However, trauma can disrupt this system, with the amygdala implicated as a mediator of these impairments in behavior. Recent evidence has further highlighted the context of infant trauma as a critical variable in determining its immediate and enduring consequences, with trauma experienced from an attachment figure, such as occurs in cases of caregiver-child maltreatment, as particularly detrimental. This review focuses on the unique role of caregiver presence during early-life trauma in programming deficits in social behavior and threat processing. Using data primarily from rodent models, we describe the interaction between trauma and attachment during a sensitive period in early life, which highlights the role of the caregiver's presence in engagement of attachment brain circuitry and suppressing threat processing by the amygdala. These data suggest that trauma experienced directly from an abusive caregiver and trauma experienced in the presence of caregiver cues produce similar neurobehavioral deficits, which are unique from those resulting from trauma alone. We go on to integrate this information into social experience throughout the lifespan, including consequences for complex scenarios, such as dominance hierarchy formation and maintenance.
Collapse
Affiliation(s)
- Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA.
| | - Elizabeth Gould
- Department of Psychology, Princeton University, Princeton, NJ, USA
| | - Regina Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, USA
| |
Collapse
|
34
|
Al Aïn S, Perry RE, Nuñez B, Kayser K, Hochman C, Brehman E, LaComb M, Wilson DA, Sullivan RM. Neurobehavioral assessment of maternal odor in developing rat pups: implications for social buffering. Soc Neurosci 2017; 12:32-49. [PMID: 26934130 PMCID: PMC5033694 DOI: 10.1080/17470919.2016.1159605] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Social support can attenuate the behavioral and stress hormone response to threat, a phenomenon called social buffering. The mother's social buffering of the infant is one of the more robust examples; yet we understand little about the neurobiology. Using a rodent model, we explore the neurobiology of social buffering by assessing neural processing of the maternal odor, a major cue controlling social buffering in rat pups. We used pups before (postnatal day (PN) 7) and after (PN14, PN23) the functional emergence of social buffering. Pups were injected with 14C 2-deoxyglucose (2-DG) and presented with the maternal odor, a control preferred odor incapable of social buffering (acetophenone), or no odor. Brains were removed, processed for autoradiography and brain areas identified as important in adult social buffering were assessed, including the amygdala basolateral complex (Basolateral Amygdala [BLA]), medial prefrontal cortex (mPFC), and anterior cingulate cortex (ACC). Results suggest dramatic changes in the processing of maternal odor. PN7 pups show mPFC and ACC activation, although PN14 pups showed no activation of the mPFC, ACC, or BLA. All brain areas assessed were recruited by PN23. Additional analysis suggests substantial changes in functional connectivity across development. Together, these results imply complex nonlinear transitions in the neurobiology of social buffering in early life that may provide insight into the changing role of the mother in supporting social buffering.
Collapse
Affiliation(s)
- Syrina Al Aïn
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Rosemarie E. Perry
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
- Neuroscience and Physiology, NYU Sackler Institute, New York University School of Medicine, New York, NY, USA
| | - Bestina Nuñez
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
| | - Kassandra Kayser
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
| | - Chase Hochman
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Elizabeth Brehman
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Miranda LaComb
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
35
|
Environmental Intervention as a Therapy for Adverse Programming by Ancestral Stress. Sci Rep 2016; 6:37814. [PMID: 27883060 PMCID: PMC5121646 DOI: 10.1038/srep37814] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Ancestral stress can program stress sensitivity and health trajectories across multiple generations. While ancestral stress is uncontrollable to the filial generations, it is critical to identify therapies that overcome transgenerational programming. Here we report that prenatal stress in rats generates a transgenerationally heritable endocrine and epigenetic footprint and elevated stress sensitivity which can be alleviated by beneficial experiences in later life. Ancestral stress led to downregulated glucocorticoid receptor and prefrontal cortex neuronal densities along with precocious development of anxiety-like behaviours. Environmental enrichment (EE) during adolescence mitigated endocrine and neuronal markers of stress and improved miR-182 expression linked to brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) regulation in stressed lineages. Thus, EE may serve as a powerful intervention for adverse transgenerational programming through microRNA-mediated regulation of BDNF and NT-3 pathways. The identification of microRNAs that mediate the actions of EE highlights new therapeutic strategies for mental health conditions and psychiatric disease.
Collapse
|
36
|
Tottenham N, Galván A. Stress and the adolescent brain: Amygdala-prefrontal cortex circuitry and ventral striatum as developmental targets. Neurosci Biobehav Rev 2016; 70:217-227. [PMID: 27473936 PMCID: PMC5074883 DOI: 10.1016/j.neubiorev.2016.07.030] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 12/16/2022]
Abstract
Adolescence is a time in development when significant changes occur in affective neurobiology. These changes provide a prolonged period of plasticity to prepare the individual for independence. However, they also render the system highly vulnerable to the effects of environmental stress exposures. Here, we review the human literature on the associations between stress-exposure and developmental changes in amygdala, prefrontal cortex, and ventral striatal dopaminergic systems during the adolescent period. Despite the vast differences in types of adverse exposures presented in his review, these neurobiological systems appear consistently vulnerable to stress experienced during development, providing putative mechanisms to explain why affective processes that emerge during adolescence are particularly sensitive to environmental influences.
Collapse
Affiliation(s)
- Nim Tottenham
- Columbia University, Department of Psychology, 1190 Amsterdam Avenue MC 5501, New York, NY 10027, United States.
| | - Adriana Galván
- University of California, Los Angeles, Department of Psychology, 1285 Franz Hall BOX 951563, Los Angeles, CA 90095-1563, United States.
| |
Collapse
|
37
|
Boulanger Bertolus J, Mouly AM, Sullivan RM. Ecologically relevant neurobehavioral assessment of the development of threat learning. Learn Mem 2016; 23:556-66. [PMID: 27634146 PMCID: PMC5026204 DOI: 10.1101/lm.042218.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/02/2016] [Indexed: 11/24/2022]
Abstract
As altricial infants gradually transition to adults, their proximate environment changes. In three short weeks, pups transition from a small world with the caregiver and siblings to a complex milieu rich in dangers as their environment expands. Such contrasting environments require different learning abilities and lead to distinct responses throughout development. Here, we will review some of the learned fear conditioned responses to threats in rats during their ontogeny, including behavioral and physiological measures that permit the assessment of learning and its supporting neurobiology from infancy through adulthood. In adulthood, odor-shock conditioning produces robust fear learning to the odor that depends upon the amygdala and related circuitry. Paradoxically, this conditioning in young pups fails to support fear learning and supports approach learning to the odor previously paired with shock. This approach learning is mediated by the infant attachment network that does not include the amygdala. During the age range when pups transition from the infant to the adult circuit (10-15 d old), pups have access to both networks: odor-shock conditioning in maternal presence uses the attachment circuit but the adult amygdala-dependent circuit when alone. However, throughout development (as young as 5 d old) the attachment associated learning can be overridden and amygdala-dependent fear learning supported, if the mother expresses fear in the presence of the pup. This social modulation of the fear permits the expression of defense reactions in life threatening situations informed by the caregiver but prevents the learning of the caregiver itself as a threat.
Collapse
Affiliation(s)
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, INSERM U1028; CNRS UMR5292; University Lyon1, Lyon, France
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York 10010, USA
| |
Collapse
|
38
|
Stockman SL, McCarthy MM. Predator odor exposure of rat pups has opposite effects on play by juvenile males and females. Pharmacol Biochem Behav 2016; 152:20-29. [PMID: 27569603 DOI: 10.1016/j.pbb.2016.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 11/17/2022]
Abstract
Juvenile social play behavior is one of the earliest sexually differentiated behaviors to emerge. In rats, as with most other species that play, males engage in more rough-and-tumble play compared to females. Exposure to early life adversity is a major driver of adult health and can manifest differently in males and females. However, the effects of adverse early life exposure on play behavior in the juvenile period are poorly understood. To address this, male and female neonatal rats were exposed to predator odor (PO), for 5min/day on PN1-PN3. At the time of exposure to PO, both male and female pups suppressed ultrasonic vocalization and displayed more freezing behavior. Circulating corticosterone increased in males immediately following PO exposure but not in females. The enduring effects of PO exposure were opposite in males compared to females in that PO exposed males decreased social play, while PO exposed females increased play behavior compared to same sex controls. PO exposure did not significantly affect cell genesis in the neonatal dentate gyrus of either sex. PO exposure did not affect anxiety-like behavior assessed in the juvenile period or in adulthood, nor did it affect social interactions in adulthood. This work provides new insight into how sex may interact with adverse early life events to contribute to development of the social consequences of such exposures.
Collapse
Affiliation(s)
- Sara L Stockman
- University of Maryland School of Medicine, 655 West Baltimore Street, Bressler Research Building 5-014, Baltimore, MD 21201, United States.
| | - Margaret M McCarthy
- University of Maryland School of Medicine, 655 West Baltimore Street, Bressler Research Building 5-014, Baltimore, MD 21201, United States.
| |
Collapse
|
39
|
Mukhin VN, Abdurasulova IN, Pavlov KI, Kozlov AP, Klimenko VM. Effects of Activation of κ-Opioid Receptors on Behavior during Postnatal Formation of the Stress Reactivity Systems. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s11055-016-0288-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Deal AL, Erickson KJ, Shiers SI, Burman MA. Limbic system development underlies the emergence of classical fear conditioning during the third and fourth weeks of life in the rat. Behav Neurosci 2016; 130:212-30. [PMID: 26820587 DOI: 10.1037/bne0000130] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Classical fear conditioning creates an association between an aversive stimulus and a neutral stimulus. Although the requisite neural circuitry is well understood in mature organisms, the development of these circuits is less well studied. The current experiments examine the ontogeny of fear conditioning and relate it to neuronal activation assessed through immediate early gene (IEG) expression in the amygdala, hippocampus, perirhinal cortex, and hypothalamus of periweanling rats. Rat pups were fear conditioned, or not, during the third or fourth weeks of life. Neuronal activation was assessed by quantifying expression of FBJ osteosarcoma oncogene (FOS) using immunohistochemistry (IHC) in Experiment 1. Fos and early growth response gene-1 (EGR1) expression was assessed using qRT-PCR in Experiment 2. Behavioral data confirm that both auditory and contextual fear continue to emerge between PD 17 and 24. The IEG expression data are highly consistent with these behavioral results. IHC results demonstrate significantly more FOS protein expression in the basal amygdala of fear-conditioned PD 23 subjects compared to control subjects, but no significant difference at PD 17. qRT-PCR results suggest specific activation of the amygdala only in older subjects during auditory fear expression. A similar effect of age and conditioning status was also observed in the perirhinal cortex during both contextual and auditory fear expression. Overall, the development of fear conditioning occurring between the third and fourth weeks of life appears to be at least partly attributable to changes in activation of the amygdala and perirhinal cortex during fear conditioning or expression. (PsycINFO Database Record
Collapse
|
41
|
Tallot L, Doyère V, Sullivan RM. Developmental emergence of fear/threat learning: neurobiology, associations and timing. GENES, BRAIN, AND BEHAVIOR 2016; 15:144-54. [PMID: 26534899 PMCID: PMC5154388 DOI: 10.1111/gbb.12261] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 02/01/2023]
Abstract
Pavlovian fear or threat conditioning, where a neutral stimulus takes on aversive properties through pairing with an aversive stimulus, has been an important tool for exploring the neurobiology of learning. In the past decades, this neurobehavioral approach has been expanded to include the developing infant. Indeed, protracted postnatal brain development permits the exploration of how incorporating the amygdala, prefrontal cortex and hippocampus into this learning system impacts the acquisition and expression of aversive conditioning. Here, we review the developmental trajectory of these key brain areas involved in aversive conditioning and relate it to pups' transition to independence through weaning. Overall, the data suggests that adult-like features of threat learning emerge as the relevant brain areas become incorporated into this learning. Specifically, the developmental emergence of the amygdala permits cue learning and the emergence of the hippocampus permits context learning. We also describe unique features of learning in early life that block threat learning and enhance interaction with the mother or exploration of the environment. Finally, we describe the development of a sense of time within this learning and its involvement in creating associations. Together these data suggest that the development of threat learning is a useful tool for dissecting adult-like functioning of brain circuits, as well as providing unique insights into ecologically relevant developmental changes.
Collapse
Affiliation(s)
- L. Tallot
- Institut des Neurosciences Paris Saclay (Neuro-PSI), UMR 9197, CNRS/Université Paris-Sud, Orsay, France
- Emotional Brain Institute, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg
- Child Study Center Institute for Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - V. Doyère
- Institut des Neurosciences Paris Saclay (Neuro-PSI), UMR 9197, CNRS/Université Paris-Sud, Orsay, France
| | - R. M. Sullivan
- Emotional Brain Institute, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg
- Child Study Center Institute for Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
42
|
Opendak M, Sullivan RM. Unique neurobiology during the sensitive period for attachment produces distinctive infant trauma processing. Eur J Psychotraumatol 2016; 7:31276. [PMID: 27837581 PMCID: PMC5106868 DOI: 10.3402/ejpt.v7.31276] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/28/2016] [Accepted: 07/31/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Trauma has neurobehavioral effects when experienced at any stage of development, but trauma experienced in early life has unique neurobehavioral outcomes related to later life psychiatric sequelae. Recent evidence has further highlighted the context of infant trauma as a critical variable in determining its immediate and enduring consequences. Trauma experienced from an attachment figure, such as occurs in cases of caregiver child maltreatment, is particularly detrimental. METHODS Using data primarily from rodent models, we review the literature on the interaction between trauma and attachment in early life, which highlights the role of the caregiver's presence in engagement of attachment brain circuitry and suppressing threat processing by the amygdala. We then consider how trauma with and without the caregiver produces long-term changes in emotionality and behavior, and suggest that these experiences initiate distinct pathways to pathology. RESULTS Together these data suggest that infant trauma processing and its enduring effects are impacted by both the immaturity of brain areas for processing trauma and the unique functioning of the early-life brain, which is biased toward processing information within the attachment circuitry. CONCLUSION An understanding of developmental differences in trauma processing as well as the critical role of the caregiver in further altering early life brain processing of trauma is important for developing age-relevant treatment and interventions.
Collapse
Affiliation(s)
- Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA;
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
43
|
Callaghan BL, Tottenham N. The Neuro-Environmental Loop of Plasticity: A Cross-Species Analysis of Parental Effects on Emotion Circuitry Development Following Typical and Adverse Caregiving. Neuropsychopharmacology 2016; 41:163-76. [PMID: 26194419 PMCID: PMC4677125 DOI: 10.1038/npp.2015.204] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/20/2022]
Abstract
Early experiences critically shape the structure and function of the brain. Perturbations in typical/species-expected early experiences are known to have profound neural effects, especially in regions important for emotional responding. Parental care is one species-expected stimulus that plays a fundamental role in the development of emotion neurocircuitry. Emerging evidence across species suggests that phasic variation in parental presence during the sensitive period of childhood affects the recruitment of emotional networks on a moment-to-moment basis. In addition, it appears that increasing independence from caregivers cues the termination of the sensitive period for environmental input into emotion network development. In this review, we examine how early parental care, the central nervous system, and behavior come together to form a 'neuro-environmental loop,' contributing to the formation of stable emotion regulation circuits. To achieve this end, we focus on the interaction of parental care and the developing amygdala-medial prefrontal cortex (mPFC) network-that is at the core of human emotional functioning. Using this model, we discuss how individual or group variations in parental independence, across chronic and brief timescales, might contribute to neural and emotional phenotypes that have implications for long-term mental health.
Collapse
Affiliation(s)
| | - Nim Tottenham
- Department of Psychology, Columbia University, New York, NY, USA
| |
Collapse
|
44
|
Jedd K, Hunt RH, Cicchetti D, Hunt E, Cowell R, Rogosch F, Toth S, Thomas KM. Long-term consequences of childhood maltreatment: Altered amygdala functional connectivity. Dev Psychopathol 2015; 27:1577-89. [PMID: 26535945 PMCID: PMC4635964 DOI: 10.1017/s0954579415000954] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Childhood maltreatment is a serious individual, familial, and societal threat that compromises healthy development and is associated with lasting alterations to emotion perception, processing, and regulation (Cicchetti & Curtis, 2005; Pollak, Cicchetti, Hornung, & Reed, 2000; Pollak & Tolley-Schell, 2003). Individuals with a history of maltreatment show altered structural and functional brain development in both frontal and limbic structures (Hart & Rubia, 2012). In particular, previous research has identified hyperactive amygdala responsivity associated with childhood maltreatment (e.g., Dannlowski et al., 2012). However, less is known about the impact of maltreatment on the relationship between the amygdala and other brain regions. The present study employed an emotion processing functional magnetic resonance imaging task to examine task-based activation and functional connectivity in adults who experienced maltreatment as children. The sample included adults with a history of substantiated childhood maltreatment (n = 33) and comparison adults (n = 38) who were well matched on demographic variables, all of whom have been studied prospectively since childhood. The maltreated group exhibited greater activation than comparison participants in the prefrontal cortex and basal ganglia. In addition, maltreated adults showed increased amygdala connectivity with the hippocampus and prefrontal cortex. The results suggest that the intense early stress of childhood maltreatment is associated with lasting alterations to frontolimbic circuitry.
Collapse
Affiliation(s)
- Kelly Jedd
- University of Minnesota, Institute of Child Development, Minneapolis, MN
| | - Ruskin H. Hunt
- University of Minnesota, Institute of Child Development, Minneapolis, MN
| | - Dante Cicchetti
- University of Minnesota, Institute of Child Development, Minneapolis, MN
- University of Rochester, Mt. Hope Family Center, Rochester, NY
| | - Emily Hunt
- University of Rochester, Mt. Hope Family Center, Rochester, NY
| | - Raquel Cowell
- University of Minnesota, Institute of Child Development, Minneapolis, MN
- St. Norbert College, De Pere, WI
| | - Fred Rogosch
- University of Rochester, Mt. Hope Family Center, Rochester, NY
| | - Sheree Toth
- University of Rochester, Mt. Hope Family Center, Rochester, NY
| | - Kathleen M. Thomas
- University of Minnesota, Institute of Child Development, Minneapolis, MN
| |
Collapse
|
45
|
Ehrlich DE, Josselyn SA. Plasticity-related genes in brain development and amygdala-dependent learning. GENES BRAIN AND BEHAVIOR 2015; 15:125-43. [PMID: 26419764 DOI: 10.1111/gbb.12255] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Learning about motivationally important stimuli involves plasticity in the amygdala, a temporal lobe structure. Amygdala-dependent learning involves a growing number of plasticity-related signaling pathways also implicated in brain development, suggesting that learning-related signaling in juveniles may simultaneously influence development. Here, we review the pleiotropic functions in nervous system development and amygdala-dependent learning of a signaling pathway that includes brain-derived neurotrophic factor (BDNF), extracellular signaling-related kinases (ERKs) and cyclic AMP-response element binding protein (CREB). Using these canonical, plasticity-related genes as an example, we discuss the intersection of learning-related and developmental plasticity in the immature amygdala, when aversive and appetitive learning may influence the developmental trajectory of amygdala function. We propose that learning-dependent activation of BDNF, ERK and CREB signaling in the immature amygdala exaggerates and accelerates neural development, promoting amygdala excitability and environmental sensitivity later in life.
Collapse
Affiliation(s)
- D E Ehrlich
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Department of Otolaryngology, NYU Langone School of Medicine, New York, NY, USA
| | - S A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
46
|
Osada K, Miyazono S, Kashiwayanagi M. The scent of wolves: pyrazine analogs induce avoidance and vigilance behaviors in prey. Front Neurosci 2015; 9:363. [PMID: 26500485 PMCID: PMC4595651 DOI: 10.3389/fnins.2015.00363] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Abstract
The common gray wolf (Canis lupus) is an apex predator located at the top of the food chain in the Northern Hemisphere. It preys on rodents, rabbits, ungulates, and many other kinds of mammal. However, the behavioral evidence for, and the chemical basis of, the fear-inducing impact of wolf urine on prey are unclear. Recently, the pyrazine analogs 2, 6-dimethylpyrazine, 2, 3, 5-trimethylpyrazine and 3-ethyl-2, 5-dimethyl pyrazine were identified as kairomones in the urine of wolves. When mice were confronted with a mixture of purified pyrazine analogs, vigilance behaviors, including freezing and excitation of neurons at the accessory olfactory bulb, were markedly increased. Additionally, the odor of the pyrazine cocktail effectively suppressed the approach of deer to a feeding area, and for those close to the feeding area elicited fear-related behaviors such as the "tail-flag," "flight," and "jump" actions. In this review, we discuss the transfer of chemical information from wolf to prey through the novel kairomones identified in wolf urine and also compare the characteristics of wolf kairomones with other predator-produced kairomones that affect rodents.
Collapse
Affiliation(s)
- Kazumi Osada
- Division of Physiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido Ishikari-Tobetsu, Japan
| | - Sadaharu Miyazono
- Department of Sensory Physiology, Asahikawa Medical University Asahikawa, Japan
| | | |
Collapse
|
47
|
Ayers LW, Asok A, Blaze J, Roth TL, Rosen JB. Changes in dam and pup behavior following repeated postnatal exposure to a predator odor (TMT): A preliminary investigation in Long-Evans rats. Dev Psychobiol 2015; 58:176-84. [PMID: 26394891 DOI: 10.1002/dev.21362] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 09/11/2015] [Indexed: 11/10/2022]
Abstract
The present study investigated whether repeated early postnatal exposure to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) alters behavioral responses to the stimulus later in life, at postnatal day (PN30). Long-Evans rat pups with their mothers were exposed for 20 min daily to TMT, water, or a noxious odor, butyric acid (BTA), during the first three weeks of life. Mothers exposed to TMT displayed more crouching and nursing behavior than those exposed to BTA, and TMT exposed pups emitted more ultrasonic vocalizations than BTA exposed pups. At PN30, rats were tested for freezing to TMT, water, or BTA. Rats exposed to TMT during the postnatal period displayed less freezing to TMT than rats exposed postnatally to water or BTA. Our data indicate that early-life experience with a predator cue has a significant impact on later fear responses to that same cue, highlighting the programming capacity of the postnatal environment on the development of behavior.
Collapse
Affiliation(s)
- Luke W Ayers
- Department of Psychology, Widener University, Chester, PA, 19013
| | - Arun Asok
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716
| | - Jennifer Blaze
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716
| | - Jeffrey B Rosen
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716.
| |
Collapse
|
48
|
Change in the hedonic value of an aversive stimulus in the presence of a pre-exposed odor. Physiol Behav 2015; 148:51-7. [DOI: 10.1016/j.physbeh.2014.12.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 12/10/2014] [Accepted: 12/13/2014] [Indexed: 11/22/2022]
|
49
|
Abstract
Intrauterine methamphetamine exposure adversely affects the neurofunctional profile of exposed children, leading to a variety of higher order cognitive deficits, such as decreased attention, reduced working-memory capability, behavioral dysregulation, and spatial memory impairments (Kiblawi et al. in J Dev Behav Pediatr 34:31-37, 2013; Piper et al. in Pharmacol Biochem Behav 98:432-439 2011; Roussotte et al. in Neuroimage 54:3067-3075, 2011; Twomey et al. in Am J Orthopsychiatry 83:64-72, 2013). In animal models of developmental methamphetamine, both neuroanatomical and behavioral outcomes critically depend on the timing of methamphetamine administration. Methamphetamine exposure during the third trimester human equivalent period of brain development results in well-defined and persistent wayfinding and spatial navigation deficits in rodents (Vorhees et al. in Neurotoxicol Teratol 27:117-134, 2005, Vorhees et al. in Int J Dev Neurosci 26:599-610, 2008; Vorhees et al. in Int J Dev Neurosci 27:289-298, 2009; Williams et al. in Psychopharmacology (Berl) 168:329-338, 2003b), whereas drug delivery during the first and second trimester equivalents produces no such effect (Acuff-Smith et al. in Neurotoxicol Teratol 18:199-215, 1996; Schutova et al. in Physiol Res 58:741-750, 2009a; Slamberova et al. in Naunyn Schmiedebergs Arch Pharmacol 380:109-114, 2009, Slamberova et al. in Physiol Res 63:S547-S558, 2014b). In this review, we examine the impact of developmental methamphetamine on emerging neural circuitry, neurotransmission, receptor changes, and behavioral outcomes in animal models. The review is organized by type of effects and timing of drug exposure (prenatal only, pre- and neonatal, and neonatal only). The findings elucidate functional patterns of interconnected brain structures (e.g., frontal cortex and striatum) and neurotransmitters (e.g., dopamine and serotonin) involved in methamphetamine-induced developmental neurotoxicity.
Collapse
|
50
|
Ryan SJ, Ehrlich DE, Rainnie DG. Morphology and dendritic maturation of developing principal neurons in the rat basolateral amygdala. Brain Struct Funct 2014; 221:839-54. [PMID: 25381464 DOI: 10.1007/s00429-014-0939-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 11/01/2014] [Indexed: 02/08/2023]
Abstract
The basolateral nucleus of the amygdala (BLA) assigns emotional valence to sensory stimuli, and many amygdala-dependent behaviors undergo marked development during postnatal life. We recently showed principal neurons in the rat BLA undergo dramatic changes to their electrophysiological properties during the first postnatal month, but no study to date has thoroughly characterized changes to morphology or gene expression that may underlie the functional development of this neuronal population. We addressed this knowledge gap with reconstructions of biocytin-filled principal neurons in the rat BLA at postnatal days 7 (P7), 14, 21, 28, and 60. BLA principal neurons underwent a number of morphological changes, including a twofold increase in soma volume from P7 to P21. Dendritic arbors expanded significantly during the first postnatal month and achieved a mature distribution around P28, in terms of total dendritic length and distance from soma. The number of primary dendrites and branch points were consistent with age, but branch points were found farther from the soma in older animals. Dendrites of BLA principal neurons at P7 had few spines, and spine density increased nearly fivefold by P21. Given the concurrent increase in dendritic material, P60 neurons had approximately 17 times as many total spines as P7 neurons. Together, these developmental transitions in BLA principal neuron morphology help explain a number of concomitant electrophysiological changes during a critical period in amygdala development.
Collapse
Affiliation(s)
- Steven J Ryan
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA
| | - David E Ehrlich
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA
| | - Donald G Rainnie
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA.
| |
Collapse
|