1
|
Lion M, Muller M, Ibrahim EC, El-Hage W, Lengvenyte A, Courtet P, Lefrere A, Belzeaux R. Role of depression, suicide attempt history and childhood trauma in neutrophil-to-lymphocyte ratio dynamics: A 30-week prospective study. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111227. [PMID: 39709173 DOI: 10.1016/j.pnpbp.2024.111227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Studying the biology of suicidal behaviour and developing blood-based biomarkers may help stratify individuals with suicidal behaviors into clinically relevant categories. Literature suggests that people diagnosed with mood disorders and suicidal behaviour show an increased neutrophil to lymphocyte ratio (NLR). For the first time, we investigated NLR variability in mood disorders, a critical aspect of biomarker development. Our study provides a result on the influence of our variables on the NLR and also on the intrinsic properties of the ratio. Consequently, our objective was to analyse the differences in NLR between healthy subjects and patients diagnosed with mood disorder with suicidal behaviour or mood disorder without suicidal attempt. A prospective study was conducted on 97 healthy subjects, 63 patients with mood disorder without suicidal behaviour and 61 patients with mood disorder with suicidal behaviour (mean age [SD] = 44.2 [14.31]; 66.1 % female). Participants were assessed four times over 30 weeks, where blood samples and clinical data were collected. After controlling for confounding factors such as smoking and medical history, we found that NLR stability was low but NLR was significantly associated with a history of suicide attempt (mixed linear model, F = 4.044; p = 0.018). We also observed a significant interaction between NLR values and childhood trauma (p = 0.002). Furthermore, our results demonstrate that NLR is influenced by childhood trauma, including in controls (p = 0.014). Finally, NLR expression differs between patients with and without suicidal behaviour, but only in those without a history of childhood trauma (p = 0.026). Despite its variability over time, our data suggest that NLR may be a promising biomarker for identifying individuals at high risk of suicidal behaviour among patients with mood disorders.
Collapse
Affiliation(s)
- M Lion
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| | - M Muller
- Department of psychiatry and Neurosciences, CERVO Brain Research centre, Université Laval, Québec City, QC G1J 2G3, Canada
| | - E C Ibrahim
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France; Fondation FondaMental, Créteil, France
| | - W El-Hage
- CHRU de Tours, Pôle de Psychiatrie et d'Addictologie, 37044 Tours, France; UMR 1253, iBraiN, INSERM, Université de Tours, 37000 Tours, France
| | - A Lengvenyte
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France; Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
| | - P Courtet
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France; Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
| | - A Lefrere
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France; Pôle de Psychiatrie, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - R Belzeaux
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France; Departement of psychiatry, CHU Montpellier, Montpellier, France.
| |
Collapse
|
2
|
Acosta H, Jansen A, Kircher T. The association between childhood adversity and hippocampal volumes is moderated by romantic relationship experiences. Eur J Neurosci 2025; 61:e16593. [PMID: 39551574 DOI: 10.1111/ejn.16593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
Reduced hippocampal volumes are a feature of many mental disorders. Childhood maltreatment is a known risk factor for the development of psychopathology and has consistently been linked to hippocampal volume reductions in adults, but not in children and adolescents. We propose that maltreatment-related difficulties in coping with developmental tasks in adolescence and young adulthood might underlie the delayed emergence of hippocampal volume reductions in maltreated individuals. In a study with 196 healthy young adults (mean age [years]: 24.0 ± 3.2, 50% female, 20.6% living with a partner (missings: n = 2)), we investigated the interaction between childhood maltreatment (Childhood Trauma Screener) and the breakup of a steady romantic relationship (List of Threatening Experiences Questionnaire) on hippocampal magnetic resonance imaging grey matter volumes. The experience of a romantic relationship breakup moderated the association between childhood maltreatment and bilateral hippocampal volumes, revealing more negative associations with hippocampal volumes in participants with at least one breakup compared to those with no breakup experience (right hippocampus: β = - 0.05 ± 0.02, p = 0.031, p (FDR) = 0.031; left hippocampus: β = -0.06 ± 0.02, p = 0.005, p (FDR) = 0.009). Moreover, our findings provide some evidence that childhood maltreatment is related to smaller bilateral hippocampal volumes only in those adults who suffered from a relationship breakup (right hippocampus: β = -0.23 ± 0.10, p = 0.018, p (FDR) = 0.018; left hippocampus: β = -0.24 ± 0.10, p = 0.016, p (FDR) = 0.018;). Our study highlights the interaction of adult social bonds with early adversity on vulnerability to psychopathology.
Collapse
Affiliation(s)
- H Acosta
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, Germany
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - A Jansen
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, Germany
- Core Facility Brainimaging, Faculty of Medicine, Philipps University Marburg, Germany
| | - T Kircher
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, Germany
| |
Collapse
|
3
|
Mundorf A, Freund N. Effects of Early Stress Exposure on Anxiety-like Behavior and MORC1 Expression in Rats. Biomolecules 2024; 14:1587. [PMID: 39766294 PMCID: PMC11674774 DOI: 10.3390/biom14121587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Exposure to stress during early and late childhood can lead to long-lasting neurobiological and behavioral impairments. Although sensitive periods for stress exposure are well established, less is known about the trajectory of induced alterations throughout development. In this study, we investigated the impact of maternal separation (MS), social isolation, and their combination on anxiety-like behavior and gene expression across developmental stages. Sprague Dawley rats were exposed to one or both stressors and later assessed for anxiety-like behavior in juvenility, adolescence, and adulthood. mRNA levels of Morc1, a gene linked to early-life stress and depression, were measured in the medial prefrontal cortex to assess developmental changes. The results showed that MS had age- and sex-dependent effects on anxiety-like behavior. Juveniles exhibited less anxiety after MS, while adolescents showed more pronounced behavioral changes following social isolation. No behavioral changes were observed in adults. Males exhibited greater anxiety-like behavior than females in adolescence and adulthood, but not in juvenility. Female adults exposed to both MS and social isolation had significantly lower Morc1 expression compared to controls. These findings highlight the dynamic effects of early stress across the lifespan, underscoring the critical role of adolescence and differential stress susceptibility by age and sex.
Collapse
Affiliation(s)
- Annakarina Mundorf
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr University Bochum, 44801 Bochum, Germany
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School, 20457 Hamburg, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
4
|
Alves J, Dos Santos APB, Vieira ADS, Martini APR, de Lima RMS, Smaniotto TÂ, de Moraes RO, Gomes RF, Acerbi GCDA, de Assis EZB, Lampert C, Dalmaz C, Couto Pereira NDS. Coping with the experience of frustration throughout life: Sex- and age-specific effects of early life stress on the susceptibility to reward devaluation. Neuroscience 2024; 553:160-171. [PMID: 38960089 DOI: 10.1016/j.neuroscience.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024]
Abstract
Early life stress may lead to lifelong impairments in psychophysiological functions, including emotional and reward systems. Unpredicted decrease in reward magnitude generates a negative emotional state (frustration) that may be involved with susceptibility to psychiatric disorders. We evaluated, in adolescents and adult rats of both sexes, whether maternal separation (MS) alters the ability to cope with an unexpected reduction of reward later in life. Litters of Wistar rats were divided into controls (non handled - NH) or subjected to MS. Animals were trained to find sugary cereal pellets; later the amount was reduced. Increased latency to reach the reward-associated area indicates higher inability to regulate frustration. The dorsal hippocampus (dHC) and basolateral amygdala (BLA) were evaluated for protein levels of NMDA receptor subunits (GluN2A/GluN2B), synaptophysin, PSD95, SNAP-25 and CRF1. We found that adult MS males had greater vulnerability to reward reduction, together with decreased GluN2A and increased GluN2B immunocontent in the dHC. MS females and adolescents did not differ from controls. We concluded that MS enhances the response to frustration in adult males. The change in the ratio of GluN2A and GluN2B subunits in dHC could be related to a stronger, more difficult to update memory of the aversive experience.
Collapse
Affiliation(s)
- Joelma Alves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Paula Bosquetti Dos Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Aline Dos Santos Vieira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Paula Rodrigues Martini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Randriely Merscher Sobreira de Lima
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Thiago Ângelo Smaniotto
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rafael Oliveira de Moraes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Roger Ferreira Gomes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Giulia Conde de Albite Acerbi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Eduardo Z B de Assis
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carine Lampert
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carla Dalmaz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Natividade de Sá Couto Pereira
- Psychological Neuroscience Laboratory, Psychology Research Centre (CIPsi), School of Psychology, University of Minho, Braga, Portugal.
| |
Collapse
|
5
|
Pifer GC, Ferrara NC, Kwapis JL. Long-lasting effects of disturbing the circadian rhythm or sleep in adolescence. Brain Res Bull 2024; 213:110978. [PMID: 38759704 PMCID: PMC11197883 DOI: 10.1016/j.brainresbull.2024.110978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Circadian rhythms are endogenous, near 24-hour rhythms that regulate a multitude of biological and behavioral processes across the diurnal cycle in most organisms. Over the lifespan, a bell curve pattern emerges in circadian phase preference (i.e. chronotype), with children and adults generally preferring to wake earlier and fall asleep earlier, and adolescents and young adults preferring to wake later and fall asleep later than their adult counterparts. This well-defined shift speaks to the variability of circadian rhythmicity over the lifespan and the changing needs and demands of the brain as an organism develops, particularly in the adolescent period. Indeed, adolescence is known to be a critical period of development during which dramatic neuroanatomical changes are occurring to allow for improved decision-making. Due to the large amount of re-structuring occurring in the adolescent brain, circadian disruptions during this period could have adverse consequences that persist across the lifespan. While the detrimental effects of circadian disruptions in adults have been characterized in depth, few studies have longitudinally assessed the potential long-term impacts of circadian disruptions during adolescence. Here, we will review the evidence that disruptions in circadian rhythmicity during adolescence have effects that persist into adulthood. As biological and social time often conflict in modern society, with school start times misaligned with adolescents' endogenous rhythms, it is critical to understand the long-term impacts of disrupted circadian rhythmicity in adolescence.
Collapse
Affiliation(s)
- Gretchen C Pifer
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Nicole C Ferrara
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Janine L Kwapis
- Department of Biology, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
6
|
Andersen SL. Increasing CB2 Receptor Activity after Early Life Stress Prevents Depressive Behavior in Female Rats. Biomolecules 2024; 14:464. [PMID: 38672480 PMCID: PMC11047932 DOI: 10.3390/biom14040464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/28/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Early adversity, the loss of the inhibitory GABAergic interneuron parvalbumin, and elevated neuroinflammation are associated with depression. Individuals with a maltreatment history initiate medicinal cannabis use earlier in life than non-maltreated individuals, suggesting self-medication. Female rats underwent maternal separation (MS) between 2 and 20 days of age to model early adversity or served as colony controls. The prelimbic cortex and behavior were examined to determine whether MS alters the cannabinoid receptor 2 (CB2), which has anti-inflammatory properties. A reduction in the CB2-associated regulatory enzyme MARCH7 leading to increased NLRP3 was observed with Western immunoblots in MS females. Immunohistochemistry with stereology quantified numbers of parvalbumin-immunoreactive cells and CB2 at 25, 40, and 100 days of age, revealing that the CB2 receptor associated with PV neurons initially increases at P25 and subsequently decreases by P40 in MS animals, with no change in controls. Confocal and triple-label microscopy suggest colocalization of these CB2 receptors to microglia wrapped around the parvalbumin neuron. Depressive-like behavior in MS animals was elevated at P40 and reduced with the CB2 agonist HU-308 or a CB2-overexpressing lentivirus microinjected into the prelimbic cortex. These results suggest that increasing CB2 expression by P40 in the prelimbic cortex prevents depressive behavior in MS female rats.
Collapse
Affiliation(s)
- Susan L Andersen
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Tomoda A, Nishitani S, Takiguchi S, Fujisawa TX, Sugiyama T, Teicher MH. The neurobiological effects of childhood maltreatment on brain structure, function, and attachment. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01779-y. [PMID: 38466395 DOI: 10.1007/s00406-024-01779-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
Childhood maltreatment is a risk factor for psychopathologies, and influences brain development at specific periods, particularly during early childhood and adolescence. This narrative review addresses phenotypic alterations in sensory systems associated with specific types of childhood maltreatment exposure, periods of vulnerability to the neurobiological effects of maltreatment, and the relationships between childhood maltreatment and brain structure, function, connectivity, and network architecture; psychopathology; and resilience. It also addresses neurobiological alterations associated with maternal communication and attachment disturbances, and uses laboratory-based measures during infancy and case-control studies to elucidate neurobiological alterations in reactive attachment disorders in children with maltreatment histories. Moreover, we review studies on the acute effects of oxytocin on reactive attachment disorder and maltreatment and methylation of oxytocin regulatory genes. Epigenetic changes may play a critical role in initiating or producing the atypical structural and functional brain alterations associated with childhood maltreatment. However, these changes could be reversed through psychological and pharmacological interventions, and by anticipating or preventing the emergence of brain alterations and subsequent psychopathological risks.
Collapse
Affiliation(s)
- Akemi Tomoda
- Research Center for Child Mental Development, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
- Division of Developmental Higher Brain Functions, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Fukui, Japan.
- Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan.
| | - Shota Nishitani
- Research Center for Child Mental Development, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
- Division of Developmental Higher Brain Functions, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Fukui, Japan
| | - Shinichiro Takiguchi
- Division of Developmental Higher Brain Functions, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Fukui, Japan
- Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan
| | - Takashi X Fujisawa
- Research Center for Child Mental Development, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
- Division of Developmental Higher Brain Functions, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Fukui, Japan
| | - Toshiro Sugiyama
- Research Center for Child Mental Development, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
- Division of Developmental Higher Brain Functions, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Fukui, Japan
- Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan
| | - Martin H Teicher
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, USA
- Department of Psychiatry, Harvard Medical School, Boston, USA
| |
Collapse
|
8
|
Rubinstein MR, Burgueño AL, Quiroga S, Wald MR, Genaro AM. Current Understanding of the Roles of Gut-Brain Axis in the Cognitive Deficits Caused by Perinatal Stress Exposure. Cells 2023; 12:1735. [PMID: 37443769 PMCID: PMC10340286 DOI: 10.3390/cells12131735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.
Collapse
Affiliation(s)
- Mara Roxana Rubinstein
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| | | | | | | | - Ana María Genaro
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| |
Collapse
|
9
|
Solarz A, Majcher-Maślanka I, Kryst J, Chocyk A. Early-life stress affects peripheral, blood-brain barrier, and brain responses to immune challenge in juvenile and adult rats. Brain Behav Immun 2023; 108:1-15. [PMID: 36400335 DOI: 10.1016/j.bbi.2022.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/21/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
Early-life stress (ELS) may affect brain maturation and neuroimmune interactions and, consequently, the inflammatory response to subsequent environmental factors later in life. Recently, the coexistence of blood-brain barrier (BBB) dysfunction and inflammation has been implicated in the etiology and progression of mental and/or neurodegenerative diseases. There are sex differences in the prevalence and outcomes of these disorders. The number of studies reporting the effects of ELS and sex on BBB functioning and neuroinflammatory processes in response to immune challenge is very limited, and the data are inconsistent. In the present study, we examined whether ELS, based on the maternal separation (MS) paradigm in rats, can condition male and female subjects to subsequent lipopolysaccharide (LPS)-induced immune challenge in juvenility or adulthood. Twenty-four hours after acute LPS injection, serum proinflammatory cytokines were measured, and BBB permeability in the medial prefrontal cortex (mPFC) and hippocampus (HP) was evaluated. Additionally, the mRNA expression of neuroinflammatory markers and BBB-related genes was also studied. We found that a single LPS challenge induced a proinflammatory response both in the periphery and in the mPFC and HP and increased BBB permeability in a sex-dependent fashion. Moreover, MS enhanced the neuroinflammatory response to LPS challenge in males (especially juveniles), whereas MS females showed no difference or a blunted central response to LPS compared with control females, mainly during adulthood. These results suggest that ELS may precondition individuals to subsequent environmental factors later in life in a sex-specific manner and potentially determine their susceptibility or resilience to mental and/or neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Joanna Kryst
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland; Department of Chemistry and Biochemistry, Institute for Basics Sciences, Faculty of Physiotherapy, University of Physical Education, Jana Pawła II Av. 78, 31-571 Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland.
| |
Collapse
|
10
|
Couto Pereira NDS, Klippel Zanona Q, Pastore Bernardi M, Alves J, Dalmaz C, Calcagnotto ME. Aversive memory reactivation: A possible role for delta oscillations in the hippocampus-amygdala circuit. J Neurosci Res 2023; 101:48-69. [PMID: 36128957 DOI: 10.1002/jnr.25127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/25/2022] [Accepted: 09/09/2022] [Indexed: 02/03/2023]
Abstract
Memory labilization, the process by which memories become susceptible to update, is essential for memory reconsolidation and has been a target for novel therapies for traumatic memory-associated disorders. Maternal separation (MS) in male rats produced memories resistant to labilization in adulthood. Based on previous results, we hypothesized that temporal desynchronization between the dorsal hippocampus (DHc) and the basolateral amygdala (BLA), during memory retrieval, could be responsible for this impairment. Our goal was to investigate possible differences in oscillatory activity and synchrony between the DHc and BLA during fear memory reactivation, between MS and non-handled (NH) rats. We used male adult Wistar rats, NH or MS, with electrodes for local field potential (LFP) recordings implanted in the DHc and BLA. Animals were submitted to aversive memory reactivation by exposure to the conditioned context (Reat) or to pseudo-reactivation in a neutral context (pReat), and LFP was recorded. Plasticity markers linked to reconsolidation were evaluated one hour after reactivation. The power of delta oscillations and DHc-BLA synchrony in Reat animals was increased, during freezing. Besides, delta modulation of gamma oscillations amplitude in the BLA was associated with the increase in DHc Zif268 levels, an immediate early gene specifically associated with reconsolidation. Concerning early life stress, we found lower power of delta and strength of delta-gamma oscillations coupling in MS rats, compared to NH, which could explain the low Zif268 levels in a subgroup of MS animals. These results suggest a role for delta oscillations in memory reactivation that should be further investigated.
Collapse
Affiliation(s)
- Natividade de Sá Couto Pereira
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Querusche Klippel Zanona
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcelo Pastore Bernardi
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Joelma Alves
- Neurobiology of Stress Laboratory, Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carla Dalmaz
- Graduate Program in Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Neurobiology of Stress Laboratory, Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
11
|
Kim EG, Chang W, Shin S, Adhikari AS, Seol GH, Song DY, Min SS. Maternal separation in mice leads to anxiety-like/aggressive behavior and increases immunoreactivity for glutamic acid decarboxylase and parvalbumin in the adolescence ventral hippocampus. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:113-125. [PMID: 36575939 PMCID: PMC9806646 DOI: 10.4196/kjpp.2023.27.1.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 12/29/2022]
Abstract
It has been reported that stressful events in early life influence behavior in adulthood and are associated with different psychiatric disorders, such as major depression, post-traumatic stress disorder, bipolar disorder, and anxiety disorder. Maternal separation (MS) is a representative animal model for reproducing childhood stress. It is used as an animal model for depression, and has well-known effects, such as increasing anxiety behavior and causing abnormalities in the hypothalamic-pituitary-adrenal (HPA) axis. This study investigated the effect of MS on anxiety or aggression-like behavior and the number of GABAergic neurons in the hippocampus. Mice were separated from their dams for four hours per day for 19 d from postnatal day two. Elevated plus maze (EPM) test, resident-intruder (RI) test, and counted glutamic acid decarboxylase 67 (GAD67) or parvalbumin (PV) positive cells in the hippocampus were executed using immunohistochemistry. The maternal segregation group exhibited increased anxiety and aggression in the EPM test and the RI test. GAD67-positive neurons were increased in the hippocampal regions we observed: dentate gyrus (DG), CA3, CA1, subiculum, presubiculum, and parasubiculum. PV-positive neurons were increased in the DG, CA3, presubiculum, and parasubiculum. Consistent with behavioral changes, corticosterone was increased in the MS group, suggesting that the behavioral changes induced by MS were expressed through the effect on the HPA axis. Altogether, MS alters anxiety and aggression levels, possibly through alteration of cytoarchitecture and output of the ventral hippocampus that induces the dysfunction of the HPA axis.
Collapse
Affiliation(s)
- Eu-Gene Kim
- Department of Anatomy and Neuroscience, Eulji University School of Medicine, Daejeon 35233, Korea
| | - Wonseok Chang
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 35233, Korea
| | - SangYep Shin
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 35233, Korea,Department of Neural Development and Disease, Korea Brain Research Institute, Daegu 41062, Korea
| | - Anjana Silwal Adhikari
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 35233, Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, Korea University School of Nursing, Seoul 02841, Korea
| | - Dae-Yong Song
- Department of Anatomy and Neuroscience, Eulji University School of Medicine, Daejeon 35233, Korea,Correspondence Dae-Yong Song, E-mail: , Sun Seek Min, E-mail:
| | - Sun Seek Min
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 35233, Korea,Correspondence Dae-Yong Song, E-mail: , Sun Seek Min, E-mail:
| |
Collapse
|
12
|
Alizadeh-Ezdini Z, Vatanparast J. Differential impact of two paradigms of early-life adversity on behavioural responses to social defeat in young adult rats and morphology of CA3 pyramidal neurons. Behav Brain Res 2022; 435:114048. [PMID: 35952779 DOI: 10.1016/j.bbr.2022.114048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/20/2022] [Accepted: 08/06/2022] [Indexed: 01/06/2023]
Abstract
Early life stress (ELS) is an important factor in programing the brain for future response to stress, and resilience or vulnerability to stress-induced emotional disorders. The hippocampal formation, with essential roles in both regulating the stress circuitry and emotionality, contributes to this adaptive programing. Here, we examined the effects of early handling (EH) and maternal deprivation (MD) as mild and intense postnatal stressors, respectively, on the behavioural responses to social defeat stress in young adulthood. We also evaluated the interaction of mild and intense ELS with later social defeat (SD) stress on the morphology and dendritic spine density of Golgi-cox-stained CA3 hippocampal neurons. SD stress in adult rats, as expected, increased anxiety and depressive-like behaviours in the open field, elevated plus-maze and forced swimming test. These effects were associated with reduction of dendritic spines and soma size of CA3 neurons. Both behavioural and structural alterations were significantly ameliorated in socially defeated rats that experienced early handling (EH-SD). Basal dendrites of CA3 neurons in EH-SD rats also showed longer dendrites and more intersections with Sholl circles in the distal portion, compared to both control and SD rats. On the other hand, in socially defeated rats with maternal deprivation experience (MD-SD) the stress-induced behavioural and structural alterations were generally intensified compared to SD rats. In MD-SD rats, apical dendrites of CA3 neurons demonstrated remarkable retraction; an effect that was not detected in SD rats. The reduction of dendritic spines density on the apical dendrites of CA3 neurons was also more pronounced in MD-SD rats compared to SD rats. Dendritic arbors and spines comprise the major neuronal substrate for the circuit connectivity, and cell region-specific alterations of dendrites and spines in CA3 neurons reveal plausible mechanisms that can underlie the impact of different ELSs on risk for affective disorders in response to social stress in adulthood.
Collapse
Affiliation(s)
| | - Jafar Vatanparast
- Department of Biology, School of Science, Shiraz University, Shiraz, Iran.
| |
Collapse
|
13
|
Keady J, Fisher M, Anderson E, LeMalenfant R, Turner J. Age-specific impacts of nicotine and withdrawal on hippocampal neuregulin signalling. Eur J Neurosci 2022; 56:4705-4719. [PMID: 35899607 PMCID: PMC9710301 DOI: 10.1111/ejn.15780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022]
Abstract
Smoking remains the leading cause of preventable death in the United States, with 87% of smokers starting before the age of 18. Age of initiation is a major predictive factor for smoking frequency and successful smoking cessation. People who initiate smoking during adolescences are 2.33 times more likely to become heavy smokers and half as likely to quit compared with smokers who started during adulthood. Additionally, schizophrenia, a disease state linked to altered neurodevelopment during adolescence, is a major predictive factor for smoking status. Smoking rates among people suffering from schizophrenia are between 60% and 90%. Interestingly, the Neuregulin Signalling Pathway (NSP), which plays an important role in neurodevelopment, is implicated in both schizophrenia and nicotine use disorder. Specifically, SNPS in neuregulin 3 (Nrg3) and Erb-B2 Receptor Tyrosine Kinase 4 (ErbB4) have been associated with smoking cessation outcomes and schizophrenia. Here, we examine the effects of chronic nicotine (18 mg/kg/day) and 24-h withdrawal on NSP gene expression in the hippocampus of adult (20-week-old) and adolescent (4-week-old) mice. We show that withdrawal from chronic nicotine decreased the expression of Erbb4 mRNA in the hippocampus of the adult mice but increased the expression of cytosolic Erbb4 protein in adolescent mice. Nrg3 mRNA and protein expression was not altered by chronic nicotine or withdrawal in the adult or adolescent cohorts, but Nrg3 mRNA and synaptosomal protein expression was lower in the adult withdrawal group when compared with their adolescent counterparts. These results highlight the age-specific effects of nicotine withdrawal on the NSP and may contribute to the lower quit rate and higher cigarette consumption of smokers who initiation during adolescences.
Collapse
Affiliation(s)
- Jack Keady
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536–0596, USA
| | - Miranda Fisher
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536–0596, USA
| | - Erin Anderson
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Rachel LeMalenfant
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Jill Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536–0596, USA
| |
Collapse
|
14
|
Lobo JJ, Ayoub LJ, Moayedi M, Linnstaedt SD. Hippocampal volume, FKBP5 genetic risk alleles, and childhood trauma interact to increase vulnerability to chronic multisite musculoskeletal pain. Sci Rep 2022; 12:6511. [PMID: 35444168 PMCID: PMC9021300 DOI: 10.1038/s41598-022-10411-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/30/2022] [Indexed: 01/05/2023] Open
Abstract
Chronic multisite musculoskeletal pain (CMP) is common and highly morbid. However, vulnerability factors for CMP are poorly understood. Previous studies have independently shown that both small hippocampal brain volume and genetic risk alleles in a key stress system gene, FKBP5, increase vulnerability for chronic pain. However, little is known regarding the relationship between these factors and CMP. Here we tested the hypothesis that both small hippocampal brain volume and FKBP5 genetic risk, assessed using the tagging risk variant, FKBP5rs3800373, increase vulnerability for CMP. We used participant data from 36,822 individuals with available genetic, neuroimaging, and chronic pain data in the UK Biobank study. Although no main effects were observed, the interaction between FKBP5 genetic risk and right hippocampal volume was associated with CMP severity (β = -0.020, praw = 0.002, padj = 0.01). In secondary analyses, severity of childhood trauma further moderated the relationship between FKBP5 genetic risk, right hippocampal brain volume, and CMP (β = -0.081, p = 0.016). This study provides novel evidence that both FKBP5 genetic risk and childhood trauma moderate the relationship between right hippocampal brain volume and CMP. The data increases our understanding of vulnerability factors for CMP and builds a foundation for further work assessing causal relationships that might drive CMP development.
Collapse
Affiliation(s)
- Jarred J Lobo
- Institute for Trauma Recovery, University of North Carolina, Campus Box #7010, Chapel Hill, NC, 27599-7010, USA
- Department of Anesthesiology, University of North Carolina, Chapel Hill, NC, USA
| | - Lizbeth J Ayoub
- Centre for Multimodal Sensorimotor and Pain Research, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- University of Toronto Centre for the Study of Pain, Toronto, ON, Canada
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Massieh Moayedi
- University of Toronto Centre for the Study of Pain, Toronto, ON, Canada.
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
- Department of Dentistry, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
- Centre for Multimodal Sensorimotor and Pain Research, Faculty of Dentistry, University of Toronto, 123 Edward Street, Suite 501B, Toronto, ON, M5G 1G6, Canada.
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, University of North Carolina, Campus Box #7010, Chapel Hill, NC, 27599-7010, USA.
- Department of Anesthesiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
15
|
Gao Y, Jiang Y, Ming Q, Zhang J, Ma R, Wu Q, Dong D, Sun X, He J, Cao W, Yuan S, Yao S. Neuroanatomical changes associated with conduct disorder in boys: influence of childhood maltreatment. Eur Child Adolesc Psychiatry 2022; 31:601-613. [PMID: 33398650 DOI: 10.1007/s00787-020-01697-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/22/2020] [Indexed: 11/30/2022]
Abstract
Childhood maltreatment (CM) poses a serious risk to the physical, emotional and psychological well-being of children, and can advance the development of maladaptive behaviors, including conduct disorder (CD). CD involves repetitive, persistent violations of others' basic rights and societal norms. Little is known about whether and how CM influences the neural mechanisms underlying CD, and CD-characteristic neuroanatomical changes have not yet been defined in a structural magnetic resonance imaging (sMRI) study. Here, we used voxel-based morphometry (VBM) and surface-based morphometry (SBM) to investigate the influence of the CD diagnosis and CM on the brain in 96 boys diagnosed with CD (62 with CM) and 86 typically developing (TD) boys (46 with CM). The participants were 12-17 years of age. Compared to the CM- CD group, the CM+ CD group had structural gray matter (GM) alterations in the fronto-limbic regions, including the left amygdala, right posterior cingulate cortex (PCC), right putamen, right dorsolateral prefrontal cortex (dlPFC) and right anterior cingulate cortex (ACC). We also found boys with CD exhibited increased GM volume in bilateral dorsomedial prefrontal cortex (dmPFC), as well as decreased GM volume and decreased gyrification in the left superior temporal gyrus (STG) relative to TD boys. Regional GM volume correlated with aggression and conduct problem severity in the CD group, suggesting that the GM changes may contribute to increased aggression and conduct problems in boys with CD who have suffered CM. In conclusion, these results demonstrate previously unreported CM-associated distinct brain structural changes among CD-diagnosed boys.
Collapse
Affiliation(s)
- Yidian Gao
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Yali Jiang
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Qingsen Ming
- Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jibiao Zhang
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Ren Ma
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Qiong Wu
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Daifeng Dong
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Xiaoqiang Sun
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Jiayue He
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Wanyi Cao
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Medical Psychological Institute of Central South University, Changsha, Hunan, China.,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China
| | - Shuwen Yuan
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuqiao Yao
- Medical Psychological Center of Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Medical Psychological Institute of Central South University, Changsha, Hunan, China. .,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, China.
| |
Collapse
|
16
|
Simone JJ, Green MR, McCormick CM. Endocannabinoid system contributions to sex-specific adolescent neurodevelopment. Prog Neuropsychopharmacol Biol Psychiatry 2022; 113:110438. [PMID: 34534603 DOI: 10.1016/j.pnpbp.2021.110438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/13/2021] [Accepted: 09/08/2021] [Indexed: 01/08/2023]
Abstract
With an increasing number of countries and states adopting legislation permitting the use of cannabis for medical purposes, there is a growing interest among health and research professionals into the system through which cannabinoids principally act, the endocannabinoid system (ECS). Much of the seminal research into the ECS dates back only 30 years and, although there has been tremendous development within the field during this time, many questions remain. More recently, investigations have emerged examining the contributions of the ECS to normative development and the effect of altering this system during important critical periods. One such period is adolescence, a unique period during which brain and behaviours are maturing and reorganizing in preparation for adulthood, including shifts in endocannabinoid biology. The purpose of this review is to discuss findings to date regarding the maturation of the ECS during adolescence and the consequences of manipulations of the ECS during this period to normative neurodevelopmental processes, as well as highlight sex differences in ECS function, important technical considerations, and future directions. Because most of what we know is derived from preclinical studies on rodents, we provide relevant background of this model and some commentary on the translational relevance of the research in this area.
Collapse
Affiliation(s)
- Jonathan J Simone
- Department of Biological Sciences, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Huxley Health Inc., 8820 Jane St., Concord, ON, L4K 2M9, Canada; eCB Consulting Inc., PO Box 652, 3 Cameron St. W., Cannington, ON L0E 1E0, Canada; Medical Cannabis Canada, 601-3500 Lakeshore Rd. W., Oakville, ON L6L 0B4, Canada.
| | - Matthew R Green
- eCB Consulting Inc., PO Box 652, 3 Cameron St. W., Cannington, ON L0E 1E0, Canada; Medical Cannabis Canada, 601-3500 Lakeshore Rd. W., Oakville, ON L6L 0B4, Canada.
| | - Cheryl M McCormick
- Department of Biological Sciences, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Department of Psychology, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
17
|
Abstract
The overarching objective is to review how early exposure to adversity interacts with inflammation to alter brain maturation. Both adversity and inflammation are significant risk factors for psychopathology. Literature relevant to the effects of adversity in children and adolescents on brain development is reviewed. These studies are supported by research in animals exposed to species-relevant stressors during development. While it is known that exposure to adversity at any age increases inflammation, the effects of inflammation are exacerbated at developmental stages when the immature brain is uniquely sensitive to experiences. Microglia play a vital role in this process, as they scavenge cellular debris and prune synapses to optimize performance. In essence, microglia modify the synapse to match environmental demands, which is necessary for someone with a history of adversity. Overall, by piecing together clinical and preclinical research areas, what emerges is a picture of how adversity uniquely sculpts the brain. Microglia interactions with the inhibitory neurotransmitter GABA (specifically, the subtype expressing parvalbumin) are discussed within contexts of development and adversity. A review of inflammation markers in individuals with a history of abuse is combined with preclinical studies to describe their effects on maturation. Inconsistencies within the literature are discussed, with a call for standardizing methodologies relating to the age of assessing adversity effects, measures to quantify stress and inflammation, and more brain-based measures of biochemistry. Preclinical studies pave the way for interventions using anti-inflammation-based agents (COX-2 inhibitors, CB2 agonists, meditation/yoga) by identifying where, when, and how the developmental trajectory goes awry.
Collapse
|
18
|
Zareie F, Ghalebandi S, Askari K, Mousavi Z, Haghparast A. Orexin receptors in the CA1 region of hippocampus modulate the stress-induced antinociceptive responses in an animal model of persistent inflammatory pain. Peptides 2022; 147:170679. [PMID: 34718063 DOI: 10.1016/j.peptides.2021.170679] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 01/01/2023]
Abstract
Stress activates multiple neural pathways and neurotransmitters that often suppress pain perception, the phenomenon called stress-induced analgesia (SIA). Orexin neurons from the lateral hypothalamus project to entire brain structures such as the hippocampus. The present study examined this hypothesis that orexinergic receptors in the CA1 region of the hippocampus may play a modulatory role in the development of SIA in formalin test as an animal model of persistent inflammatory pain. One hundred-two adult male Wistar rats were administered with intra-CA1 orexin-1 receptor (OX1r) antagonist, SB334867, at the doses of 3, 10, 30, and 100 nmol or TCS OX2 29 as orexin-2 receptor (OX2r) antagonist at the doses of 1, 3, 10, and 30 nmol. Five min later, rats were exposed to forced swim stress (FSS) for a 6-min period. Then, pain-related behaviors induced by formalin injection were measured at the 5-min blocks during a 60-min period of formalin test. The current study indicated that solely stress exposure elicits antinociception in the early and late phases of the formalin test. The FSS-induced analgesia was prevented by intra-CA1 administration of SB334867 or TCS OX2 29 during either phase of the formalin test. Moreover, the contribution of the OX2r in the mediation of analgesic effect of stress was more prominent than that of the OX1r during both phases of the formalin test. It is suggested that OX1r and OX2r in the CA1 region of the hippocampus are involved in stress-induced analgesia in the animal model of persistent inflammatory pain.
Collapse
Affiliation(s)
- Fatemeh Zareie
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyedehdelaram Ghalebandi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| | - Kobra Askari
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Zahra Mousavi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Solarz A, Majcher-Maślanka I, Chocyk A. Effects of early-life stress and sex on blood-brain barrier permeability and integrity in juvenile and adult rats. Dev Neurobiol 2021; 81:861-876. [PMID: 34320279 DOI: 10.1002/dneu.22846] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
Early-life stress (ELS) is considered a relevant etiological factor for neurodegenerative and mental disorders. In the present study, we hypothesized that ELS may persistently and sex dependently influence blood-brain barrier (BBB) integrity and function during critical periods of brain development and consequently determine susceptibility to and sex-related prevalence of chronic diseases in adult life. We used the maternal separation (MS) procedure in rats to model ELS and evaluated BBB permeability and gene expression of selected tight junction (TJ) proteins, glucose transporter type 1 (Slc2a1) and aquaporin 4 (Aqp4) in the medial prefrontal cortex (mPFC), dorsal striatum (dSTR) and hippocampus of juvenile and adult rats. Serum concentrations of a peripheral marker of BBB function (S100β) and proinflammatory cytokines were also assessed. We observed developmental sealing of the BBB and sex differences in the permeability of the BBB and the mRNA expression of TJ proteins and Slc2a1. Adult females showed lower BBB permeability and higher levels of Cldn3, Cldn5, Ocln, and Slc2a1 in the mPFC and dSTR than males. MS temporarily increased BBB permeability in the dSTR of juvenile males and affected mRNA expression of the majority of studied proteins related to BBB function in age-, region- and sex-dependent manners. Additionally, MS sex dependently decreased serum S100β levels and did not affect proinflammatory cytokine concentrations. In general, our study did not reveal a clear or strong negative effect of MS on BBB integrity. However, the results suggest that ELS may induce adaptive/maladaptive changes or compensatory mechanisms within the BBB of unknown yet consequences.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
20
|
Shirenova SD, Khlebnikova NN, Krupina NA. Long-Term Social Isolation Reduces Expression of the BDNF Precursor and Prolyl Endopeptidase in the Rat Brain. BIOCHEMISTRY (MOSCOW) 2021; 86:704-715. [PMID: 34225593 DOI: 10.1134/s0006297921060080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Early-life stress is a risk factor for the development of behavioral and cognitive disorders in humans and animals. Such stressful situations include social isolation in early postnatal ontogenesis. Behavioral and cognitive impairments associated with neuroplastic changes in brain structures. We have found that after ten weeks of social isolation, male Wistar rats show behavioral abnormalities and cognitive deficit, accompanied by an increase in the relative expression of gene encoding serine protease prolyl endopeptidase (PREP, EC 3.4.21.26) in the brain frontal cortex. The present study aimed to assess synaptophysin (SYP), brain-derived neurotrophic factor precursor (proBDNF), and PREP expression using Western blot in the brain structures - the hippocampus, frontal cortex, and striatum of the rats subjected to prolonged social isolation compared with group-housed animals. Twenty Wistar rats were used for this study (10 males and 10 females). Experimental animals (5 males and 5 females) were kept one per cage for nine months, starting from the age of one month. Ten-month-old socially isolated rats showed memory deficit in passive avoidance paradigm and Morris Water Maze and reactivity to novelty reduction. We used monoclonal antibodies for the Western blot analysis of the expression of SYP, proBDNF, and PREP in the rat brain structures. Social isolation caused a proBDNF expression reduction in the frontal cortex in females and a reduction in PREP expression in the striatum in males. These data suppose that neurotrophic factors and PREP are involved in the mechanisms of behavioral and cognitive impairments observed in the rats subjected to prolonged social isolation with an early life onset.
Collapse
Affiliation(s)
- Sofie D Shirenova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia.
| | - Nadezhda N Khlebnikova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia
| | - Nataliya A Krupina
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia
| |
Collapse
|
21
|
Conley MI, Skalaban LJ, Rapuano KM, Gonzalez R, Laird AR, Dick AS, Sutherland MT, Watts R, Casey B. Altered hippocampal microstructure and function in children who experienced Hurricane Irma. Dev Psychobiol 2021; 63:864-877. [PMID: 33325561 PMCID: PMC8206237 DOI: 10.1002/dev.22071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/31/2020] [Accepted: 11/25/2020] [Indexed: 01/12/2023]
Abstract
Hurricane Irma was the most powerful Atlantic hurricane in recorded history, displacing 6 million and killing over 120 people in the state of Florida alone. Unpredictable disasters like Irma are associated with poor cognitive and health outcomes that can disproportionately impact children. This study examined the effects of Hurricane Irma on the hippocampus and memory processes previously related to unpredictable stress. We used an innovative application of an advanced diffusion-weighted imaging technique, restriction spectrum imaging (RSI), to characterize hippocampal microstructure (i.e., cell density) in 9- to 10-year-old children who were exposed to Hurricane Irma relative to a non-exposed control group (i.e., assessed the year before Hurricane Irma). We tested the hypotheses that the experience of Hurricane Irma would be associated with decreases in: (a) hippocampal cellularity (e.g., neurogenesis), based on known associations between unpredictable stress and hippocampal alterations; and (b) hippocampal-related memory function as indexed by delayed recall. We show an association between decreased hippocampal cellularity and delayed recall memory in children who experienced Hurricane Irma relative to those who did not. These findings suggest an important role of RSI for assessing subtle microstructural changes related to functionally significant changes in the developing brain in response to environmental events.
Collapse
Affiliation(s)
- May I. Conley
- Department of PsychologyYale UniversityNew HavenCTUSA
| | | | | | - Raul Gonzalez
- Department of PsychologyFlorida International UniversityMiamiFLUSA
| | - Angela R. Laird
- Department of PhysicsFlorida International UniversityMiamiFLUSA
| | | | | | - Richard Watts
- Department of PsychologyYale UniversityNew HavenCTUSA
| | - B.J. Casey
- Department of PsychologyYale UniversityNew HavenCTUSA
| |
Collapse
|
22
|
Moreno H, de Brugada I. Prenatal dietary choline supplementation modulates long-term memory development in rat offspring. Nutr Neurosci 2021; 24:417-425. [PMID: 31304891 DOI: 10.1080/1028415x.2019.1641294] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background: Previous studies on preclinical models have shown that giving supplemental choline during the embryonic period improves performance on memory tasks during adulthood. However, the effects of an early intervention on the development of cognitive functions in the immature brain have not been widely studied. In addition, it has been well established that short-term memory in rats emerges at an earlier stage than long-term memory.Objective: The aim of this work was to examine the effect of prenatal dietary choline supplementation on long-term memory development in rats.Methods: In order to assess long-term memory, we used an object-recognition task, which evaluates the ability to recall a previously presented stimulus. Pregnant rats were fed with the diets AIN 76-A standard (1.1 g choline/Kg food) or supplemented (5 g choline/Kg food) between embryonic days (E) 12 and E18. On the first post-natal day (PN 0), male offspring of the rats fed with the supplemented and standard diet were cross-fostered to rat dams fed a standard diet during pregnancy and tested at the age of PN21-22 or PN29-31 applying 24-hour retention tests.Results: The supplemented animals spent less time exploring the familiar object after a 24-hour retention interval, an effect that was observed in both the group tested at PN21-22 days of age and that tested at PN29-31 days. The non-supplemented rats only showed this effect in the group tested at PN29-31 days.Conclusions: These results suggest that prenatal supplementation with choline accelerates the development of long-term memory in rats.
Collapse
Affiliation(s)
- Hayarelis Moreno
- Department of Psychology of Education and Psychobiology, International University of La Rioja, La Rioja, Spain
| | - Isabel de Brugada
- Department of Experimental Psychology, University of Granada, Granada, Spain
- Mind, Brain and Behavior Research Center (CIMCYC), University of Granada, Granada, Spain
| |
Collapse
|
23
|
Fowler CH, Bogdan R, Gaffrey MS. Stress-induced cortisol response is associated with right amygdala volume in early childhood. Neurobiol Stress 2021; 14:100329. [PMID: 33997154 PMCID: PMC8102621 DOI: 10.1016/j.ynstr.2021.100329] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/18/2021] [Accepted: 04/12/2021] [Indexed: 01/26/2023] Open
Abstract
Rodent research suggests that dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and the resulting cortisol stress response can alter the structure of the hippocampus and amygdala. Because early-life changes in brain structure can produce later functional impairment and potentially increase risk for psychiatric disorder, it is critical to understand the relationship between the cortisol stress response and brain structure in early childhood. However, no study to date has characterized the concurrent association between cortisol stress response and hippocampal and amygdala volume in young children. In the present study, 42 young children (M age = 5.97, SD = 0.76), completed a frustration task and cortisol response to stress was measured. Children also underwent magnetic resonance imaging (MRI), providing structural scans from which their hippocampal and amygdala volumes were extracted. Greater cortisol stress response was associated with reduced right amygdala volume, controlling for whole brain volume, age, sex, and number of cortisol samples. There were no significant associations between cortisol stress response and bilateral hippocampus or left amygdala volumes. The association between right amygdala volume and cortisol stress response raises the non-mutually exclusive possibilities that the function of the HPA axis may shape amygdala structure and/or that amygdala structure may shape HPA axis function. As both cortisol stress response and amygdala volume have been associated with risk for psychopathology, it is possible that the relationship between cortisol stress response and amygdala volume is part of a broader pathway contributing to psychiatric risk.
Collapse
Affiliation(s)
- Carina H. Fowler
- Department of Psychology & Neuroscience, Duke University, Reuben-Cooke Building, 417 Chapel Drive, Durham, NC, 27708, USA
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Somers Family Hall, Forsyth Blvd, St. Louis, Missouri, 63105, USA
| | - Michael S. Gaffrey
- Department of Psychology & Neuroscience, Duke University, Reuben-Cooke Building, 417 Chapel Drive, Durham, NC, 27708, USA
| |
Collapse
|
24
|
Bremner JD, Hoffman M, Afzal N, Cheema FA, Novik O, Ashraf A, Brummer M, Nazeer A, Goldberg J, Vaccarino V. The environment contributes more than genetics to smaller hippocampal volume in Posttraumatic Stress Disorder (PTSD). J Psychiatr Res 2021; 137:579-588. [PMID: 33168198 PMCID: PMC8345282 DOI: 10.1016/j.jpsychires.2020.10.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Studies using structural magnetic resonance imaging (MRI) volumetrics showed smaller hippocampal volume in patients with post-traumatic stress disorder (PTSD). These studies were cross-sectional and did not address whether smaller volume is secondary to stress-induced damage, or whether pre-existing factors account for the findings. The purpose of this study was to use a co-twin case control design to assess the relative contribution of genetic and environmental factors to hippocampal volume in PTSD. METHODS Monozygotic (N = 13 pairs) and dizygotic (N = 21 pairs) twins with a history of Vietnam Era military service, where one brother went to Vietnam and developed PTSD, while his brother did not go to Vietnam or develop PTSD, underwent MR imaging of the brain. Structural MRI scans were used to manually outline the left and right hippocampus on multiple coronal slices, add the areas and adjust for slice thickness to determine hippocampal volume. RESULTS Twins with Vietnam combat-related PTSD had a mean 11% smaller right hippocampal volume in comparison to their twin brothers without combat exposure or PTSD (p < .05). There was no significant interaction by zygosity, suggesting that this was not a predisposing risk factor or genetic effect. CONCLUSIONS These findings are consistent with smaller hippocampal volume in PTSD, and suggest that the effects are primarily due to environmental effects such as the stress of combat.
Collapse
Affiliation(s)
- J. Douglas Bremner
- Departments of Psychiatry and Behavioral Sciences, USA, Radiology, and Medicine (Cardiology), USA, Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA, Corresponding author. Dept of Psychiatry & Behavioral Sciences, Emory University School of Medicine, 12 Executive Park Dr NE, USA. (J.D. Bremner)
| | | | - Nadeem Afzal
- Departments of Psychiatry and Behavioral Sciences, USA
| | - Faiz A. Cheema
- Departments of Psychiatry and Behavioral Sciences, USA, The Vietnam Era Twin Registry, Seattle Veterans Administration Epidemiology Research, USA
| | - Olga Novik
- Departments of Psychiatry and Behavioral Sciences, USA, The Vietnam Era Twin Registry, Seattle Veterans Administration Epidemiology Research, USA
| | - Ali Ashraf
- Departments of Psychiatry and Behavioral Sciences, USA
| | | | - Ahsan Nazeer
- Departments of Psychiatry and Behavioral Sciences, USA
| | - Jack Goldberg
- Information Center and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Viola Vaccarino
- Emory University School of Medicine, Atlanta GA; Atlanta VAMC, Decatur, GA, USA, The Vietnam Era Twin Registry, Seattle Veterans Administration Epidemiology Research, USA
| |
Collapse
|
25
|
Sun H, Zhang X, Kong Y, Gou L, Lian B, Wang Y, Jiang L, Li Q, Sun H, Sun L. Maternal Separation-Induced Histone Acetylation Correlates with BDNF-Programmed Synaptic Changes in an Animal Model of PTSD with Sex Differences. Mol Neurobiol 2021; 58:1738-1754. [PMID: 33245480 DOI: 10.1007/s12035-020-02224-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Maternal separation (MS) causes long-lasting epigenetic changes in the brain and increases vulnerability to traumatic events in adulthood. Of interest, there may be sex-specific differences in these epigenetic changes. In this study, the extent of histone acetylation in the hippocampus (HIP) and the expression of BDNF were measured to determine whether BDNF influences risk of PTSD following MS in early life. Rat offspring were separated from their dams (3 h/day or 6 h/day from PND2~PND14). Then, pups were treated with a single prolonged stress (SPS) procedure when they reached adulthood (PND80). In animals stressed with the SPS procedure in adulthood, those that had increased MS intensity in childhood demonstrated more significant changes in performance on tests of anxiety, depression, and contextual fear memory. Reduced levels of total BDNF mRNA and protein were observed after SPS treatment and further declined in groups with greater MS time in childhood. Interestingly, these changes were correlated with decreased H3K9ac levels and increased HDAC2 levels. Additional MS also led to more severe ultrastructural synaptic damage in rats that experienced the SPS procedure, particularly in the CA1 and CA3 region of the HIP, reflecting impaired synaptic plasticity in these regions. Interestingly, male rats in the MS3h-PTSD group showed decreased anxiety, but no similar changes were found in female rats, suggesting a degree of gender specificity in coping with stress after mild MS. In summary, this study suggests that the epigenetic signatures of the BDNF genes can be linked to HIP responses to stress, providing insights that may be relevant for people at risk of stress-related psychopathologies.
Collapse
MESH Headings
- Acetylation
- Animals
- Behavior, Animal
- Brain-Derived Neurotrophic Factor/metabolism
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/ultrastructure
- CA3 Region, Hippocampal/metabolism
- CA3 Region, Hippocampal/ultrastructure
- Corticosterone/blood
- Disease Models, Animal
- Elevated Plus Maze Test
- Fear
- Female
- Gene Expression Regulation
- Histone Deacetylase 2/genetics
- Histone Deacetylase 2/metabolism
- Histones/metabolism
- Immobilization
- Male
- Maternal Deprivation
- Open Field Test
- Rats, Sprague-Dawley
- Regression Analysis
- Sex Characteristics
- Stress Disorders, Post-Traumatic/blood
- Stress Disorders, Post-Traumatic/complications
- Stress Disorders, Post-Traumatic/metabolism
- Stress, Psychological/blood
- Stress, Psychological/complications
- Swimming
- Synapses/metabolism
- Synapses/ultrastructure
- Rats
Collapse
Affiliation(s)
- Haoran Sun
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Xianqiang Zhang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, 100083, People's Republic of China
| | - Yujia Kong
- School of Public Health, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Luping Gou
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Yanyu Wang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Li Jiang
- Cerebral Center, Sunshine Union Hospital, 9000# Yingqian Street, Weifang, 261205, Shandong, People's Republic of China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, People's Republic of China
| | - Hongwei Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China
| | - Lin Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, 261053, Shandong, People's Republic of China.
| |
Collapse
|
26
|
Bortolotto I, de Brum APS, Guecheva TN, de Souza LM, de Paula-Ramos ALL, Trindade C, Consiglio AR. DNA damage, salivary cortisol levels, and cognitive parameters in a nursing team. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2021; 861-862:503300. [PMID: 33551101 DOI: 10.1016/j.mrgentox.2020.503300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
In a cross-sectional study of women in a nursing team at a university hospital in southern Brazil, we studied DNA damage, salivary cortisol levels, and cognition. DNA damage was measured in blood leukocytes with the comet assay and the micronucleus test. Salivary cortisol levels were determined upon waking, 30 min later, and at bedtime. Cognition was evaluated according to the Stroop, Digit span and Word span tests. Cortisol levels on waking up were associated negatively with the number of years the employee worked at the institution and positively with the DNA damage in comet assay. Cognitive scores were lower when the cortisol levels were low at awakening and high at bedtime; and were associated positively with educational level. Cortisol status may influence overall health as well as essential work skills, such as attention.
Collapse
Affiliation(s)
- Iranez Bortolotto
- Programa de Pós-Graduação em Enfermagem, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Ana Paula Scherer de Brum
- Programa de Pós-Graduação em Enfermagem, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Universidade do Oeste de Santa Catarina (UNOESC), SC, Brazil
| | - Temenouga Nikolova Guecheva
- Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Larissa Milano de Souza
- Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Ana Ligia Lia de Paula-Ramos
- Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Cristiano Trindade
- Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil; Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia.
| | - Angelica Rosat Consiglio
- Programa de Pós-Graduação em Enfermagem, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil.
| |
Collapse
|
27
|
Babicola L, Ventura R, D'Addario SL, Ielpo D, Andolina D, Di Segni M. Long term effects of early life stress on HPA circuit in rodent models. Mol Cell Endocrinol 2021; 521:111125. [PMID: 33333214 DOI: 10.1016/j.mce.2020.111125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 01/06/2023]
Abstract
Adaptation to environmental challenges represents a critical process for survival, requiring the complex integration of information derived from both external cues and internal signals regarding current conditions and previous experiences. The Hypothalamic-pituitary-adrenal axis plays a central role in this process inducing the activation of a neuroendocrine signaling cascade that affects the delicate balance of activity and cross-talk between areas that are involved in sensorial, emotional, and cognitive processing such as the hippocampus, amygdala, Prefrontal Cortex, Ventral Tegmental Area, and dorsal raphe. Early life stress, especially early critical experiences with caregivers, influences the functional and structural organization of these areas, affects these processes in a long-lasting manner and may result in long-term maladaptive and psychopathological outcomes, depending on the complex interaction between genetic and environmental factors. This review summarizes the results of studies that have modeled this early postnatal stress in rodents during the first 2 postnatal weeks, focusing on the long-term effects on molecular and structural alteration in brain areas involved in Hypothalamic-pituitary-adrenal axis function. Moreover, a brief investigation of epigenetic mechanisms and specific genetic targets mediating the long-term effects of these early environmental manipulations and at the basis of differential neurobiological and behavioral effects during adulthood is provided.
Collapse
Affiliation(s)
- Lucy Babicola
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Rossella Ventura
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Sebastian Luca D'Addario
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Donald Ielpo
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Diego Andolina
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
28
|
Li WC, Chao HT, Lin MW, Shen HD, Chen LF, Hsieh JC. Neuroprotective effect of Val variant of BDNF Val66Met polymorphism on hippocampus is modulated by the severity of menstrual pain. NEUROIMAGE-CLINICAL 2021; 30:102576. [PMID: 33561695 PMCID: PMC7873439 DOI: 10.1016/j.nicl.2021.102576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 12/19/2022]
Abstract
Primary dysmenorrhea (PDM) refers to menstrual pain of which the pathological cause(s) are unknown. This study examined the associations among BDNF Val66Met polymorphisms, menstrual pain severity, and hippocampal volume among young PDM subjects. We recruited 115 PDM subjects, including severe cases (n = 66) and moderate cases (n = 44), and 117 young females (aged 20-30 years) as a control group (CON) for BDNF Val66Met genotyping and MRI examination. The assessment of hippocampal volume involved analysis at various anatomical resolutions, i.e., whole hippocampal volume, hippocampal subfields, and voxel-based morphometry (VBM) volumetric analysis. Two-way ANOVA analyses with planned contrasts and Bonferroni correction were conducted for the assessment of hippocampal volume. Linear regression was used to test for BDNF Val66Met Val allele dosage-dependent effects. We observed no main effects of group, genotype, or group-genotype interactions on bilateral whole hippocampal volumes. Significant interactions between PDM severity and BDNF Val66Met genotype were observed in the right whole hippocampus, subiculum, and molecular layer. Post-hoc analysis revealed that the average hippocampal volume of Val/Val moderate PDM subjects was greater than that of Val/Val severe PDM subjects. Note that right hippocampal volume was greater in the Val/Val group than in the Met/Met group, particularly in the right posterior hippocampal region. Dosage effect analysis revealed a positive dosage-dependent relationship between the Val allele and volume of the right whole hippocampus, subiculum, molecular layer, and VBM-defined right posterior hippocampal region in the moderate PDM subgroup only. These findings indicate that Val/Val PDM subjects are resistant to intermittent moderate pain-related stress, whereas Met carrier PDM subjects are susceptible. When confronted with years of repeated PDM stress, the hippocampus can undergo differential structural changes in accordance with the BDNF genotype and pain severity. This triad study on PDM (i.e., combining genotype with endophenotype imaging results and clinical phenotypes), underscores the potential neurobiological consequences of PDM, which may prefigure in neuroimaging abnormalities associated with various chronic pain disorders. Our results provide evidence for Val allele dosage-dependent protective effects on the hippocampal structure; however, in cases of the Val variant, these effects were modulated in accordance with the severity of menstrual pain.
Collapse
Affiliation(s)
- Wei-Chi Li
- Institute of Brain Science, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Integrated Brain Research Unit, Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiang-Tai Chao
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Wei Lin
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Horng-Der Shen
- Laboratory of Microbiology, Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Fen Chen
- Institute of Brain Science, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Integrated Brain Research Unit, Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Jen-Chuen Hsieh
- Institute of Brain Science, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Integrated Brain Research Unit, Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
29
|
Neural meaning making, prediction, and prefrontal-subcortical development following early adverse caregiving. Dev Psychopathol 2021; 32:1563-1578. [PMID: 33427163 DOI: 10.1017/s0954579420001169] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Early adversities that are caregiving-related (crEAs) are associated with a significantly increased risk for mental health problems. Recent neuroscientific advances have revealed alterations in medial prefrontal cortex (mPFC)-subcortical circuitry following crEAs. While this work has identified alterations in affective operations (e.g., perceiving, reacting, controlling, learning) associated with mPFC-subcortical circuitry, this circuitry has a much broader function extending beyond operations. It plays a primary role in affective meaning making, involving conceptual-level, schematized knowledge to generate predictions about the current environment. This function of mPFC-subcortical circuitry motivates asking whether mPFC-subcortical phenotypes following crEAs support semanticized knowledge content (or the concept-level knowledge) and generate predictive models. I present a hypothesis motivated by research findings across four different lines of work that converge on mPFC-subcortical neuroanatomy, including (a) the neurobiology supporting emotion regulation processes in adulthood, (b) the neurobiology that is activated by caregiving cues during development, (c) the neurobiology that is altered by crEAs, and (d) the neurobiology of semantic-based meaning making. I hypothesize that the affective behaviors following crEAs result in part from affective semantic memory processes supported by mPFC-subcortical circuitry that over the course of development, construct affective schemas that generate meaning making and guide predictions. I use this opportunity to review some of the literature on mPFC-subcortical circuit development following crEAs to illustrate the motivation behind this hypothesis. Long recognized by clinical science and cognitive neuroscience, studying schema-based processes may be particularly helpful for understanding how affective meaning making arises from developmental trajectories of mPFC-subcortical circuitry.
Collapse
|
30
|
Chowdhury TG, Fenton AA, Aoki C. Effects of adolescent experience of food restriction and exercise on spatial learning and open field exploration of female rats. Hippocampus 2020; 31:170-188. [PMID: 33146453 DOI: 10.1002/hipo.23275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/29/2020] [Accepted: 10/11/2020] [Indexed: 11/08/2022]
Abstract
The hippocampus carries out multiple functions: spatial cognition dorsally (DH) and regulation of emotionality-driven behavior ventrally (VH). Previously, we showed that dendrites of DH and VH pyramidal neurons of female rats are still developing robustly during adolescence and are altered by the experience of food restriction and voluntary exercise on a wheel. We tested whether such anatomical changes during adolescence impact anxiety-like behavior and spatial cognition. Four groups of female rats were evaluated for these behaviors: those with wheel access in its cage from postnatal day (P) 36-44 (EX); those with food access restricted to 1 hr per day, from P40 to 44 (FR); those with EX from P36 to 44, combined with FR from P40 to 44, which we will refer to as EX + FR; and controls, CON (no EX, no FR). Open field test for anxiety-like behavior and active place avoidance test for spatial cognition were conducted at P47-49, the age when food restricted animals have restored body weight, or at P54-56, to identify more enduring effects. Anxiety-like behavior was elevated for the EX and FR groups at P47-49 but not for the EX + FR group. By P54-56, the EX + FR and EX groups exhibited less anxiety-like behavior, indicating a beneficial delayed main effect of exercise. There was a beneficial main effect of food restriction upon cognition, as the FR group showed cognition superior to CONs' at P44-46 and P54-56, while the EX + FR animals also showed enhanced spatial learning at P54-56. EX + FR animals with best adaptation to the feeding schedule showed the best spatial learning performance but with a delay. The EX group exhibited only a transient improvement. These findings indicate that FR, EX, and EX + FR in mid-adolescence are all beneficial in reducing anxiety-like behavior and improving spatial cognition but with subtle differences in the timing of their manifestation, possibly reflecting the protracted maturation of the hippocampus.
Collapse
Affiliation(s)
- Tara G Chowdhury
- Center for Neural Science, New York University, New York, New York, USA
| | - André A Fenton
- Center for Neural Science, New York University, New York, New York, USA
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, New York, USA
| |
Collapse
|
31
|
De Guzman RM, Medina J, Saulsbery AI, Workman JL. Rotated nursing environment with underfeeding: A form of early-life adversity with sex- and age-dependent effects on coping behavior and hippocampal neurogenesis. Physiol Behav 2020; 225:113106. [PMID: 32717197 DOI: 10.1016/j.physbeh.2020.113106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/07/2020] [Accepted: 07/24/2020] [Indexed: 01/06/2023]
Abstract
We investigated how a unique form of early-life adversity (ELA), caused by rotated nursing environment to induce underfeeding, alters anxiety-like and stress-coping behaviors in male and female Sprague Dawley rats in adolescence and adulthood. Adult female rats underwent either thelectomy (thel; surgical removal of teats), sham surgery, or no surgery (control) before mating. Following parturition, litters were rotated between sham and thel rats every 12 h to generate a group of rats that experienced ELA (rotated housing, rotated mother, and 50% food restriction) from postnatal day 0 to 26. Control litters remained with their natal, nursing dams. Regardless of age and sex, ELA reduced activity in the periphery of the open field. ELA increased immobility in the forced swim test, particularly in adults. We used doublecortin immunohistochemistry to identify immature neurons in the hippocampus. ELA increased the number and density of immature neurons in the dentate gyrus of adolescent males (but not females) and reduced the density of immature neurons in adult males (but not females). This research indicates that a unique form of ELA alters stress-related passive coping and hippocampal neurogenesis in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Angela I Saulsbery
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222, United States.
| |
Collapse
|
32
|
Mareckova K, Miles A, Andryskova L, Brazdil M, Nikolova YS. Temporally and sex-specific effects of maternal perinatal stress on offspring cortical gyrification and mood in young adulthood. Hum Brain Mapp 2020; 41:4866-4875. [PMID: 33010202 PMCID: PMC7643354 DOI: 10.1002/hbm.25163] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/28/2020] [Indexed: 01/19/2023] Open
Abstract
Maternal stress during pregnancy and shortly thereafter is associated with altered offspring brain development that may increase risk of mood and anxiety disorders. Cortical gyrification is established during the prenatal period and the first 2 years of life and is altered in psychiatric disorders. Here, we sought to characterize the effects of perinatal stress exposure on offspring gyrification patterns and mood dysregulation in young adulthood. Participants included 85 young adults (56.5% women; 23–24 years) from the European Longitudinal Study of Pregnancy and Childhood (ELSPAC) with perinatal stress data across four distinct timepoints and structural MRI data from young adulthood. Perinatal stress exposure was measured as maternal stress during first and second half of pregnancy, first 6 months, and 6–18 months after birth. Cortical gyrification and mood dysregulation were quantified using local gyrification index (LGI), computed with Freesurfer, and the Profile of Mood States questionnaire, respectively. Perinatal stress predicted cortical gyrification in young adulthood, and its timing influenced location, direction, and sex‐specificity of effects. In particular, whereas early prenatal stress was associated with sex‐dependent medium‐to‐large effects in large temporal, parietal, and occipital regions (f2 = 0.19–0.38, p < .001), later perinatal stress was associated with sex‐independent small‐to‐medium effects in smaller, more anterior regions (f2 = 0.10–0.19, p < .003). Moreover, in females, early prenatal stress predicted higher LGI in a large temporal region, which was further associated with mood disturbance in adulthood (r = 0.399, p = .006). These findings point out the long‐term implications of perinatal stress exposure for cortical morphology and mood dysregulation.
Collapse
Affiliation(s)
- Klara Mareckova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Amy Miles
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Lenka Andryskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Milan Brazdil
- Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Grigoruta M, Chavez-Solano M, Varela-Ramirez A, Sierra-Fonseca JA, Orozco-Lucero E, Hamdan JN, Gosselink KL, Martinez-Martinez A. Maternal separation induces retinal and peripheral blood mononuclear cell alterations across the lifespan of female rats. Brain Res 2020; 1749:147117. [PMID: 32971085 DOI: 10.1016/j.brainres.2020.147117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022]
Abstract
Early life stress alters the function and feedback regulation of the hypothalamic-pituitaryadrenal (HPA) axis, and can contribute to neuroinflammation and neurodegeneration by modifying peripheral blood mononuclear cell (PBMC) activity. The retina, as part of the nervous system, is sensitive to immune changes induced by stress. However, the consequences of stress experienced at an early age on retinal development have not yet been elucidated. Here we aimed to evaluate the impact of maternal separation (MatSep) across three stages of the lifespan (adolescent, adult, and aged) on the retina, as well as on progression through the cell cycle and mitochondrial activity in PBMCs from female Wistar rats. Newborn pups were separated from their mother from postnatal day (PND) 2 until PND 14 for 3 h/day. Retinal analysis from the MatSep groups showed architectural alterations such as a diminished thickness of retinal layers, as well as increased expression of proinflammatory markers DJ-1, Iba-1, and CD45 and the gliotic marker GFAP. Additionally, MatSep disrupted the cell cycle and caused long-term increases in mitochondrial activity in PBMCs from adolescent and adult rats. Changes in the cell cycle profile of the PBMCs from aged MatSep rats were undetected. However, these PBMCs exhibited increased sensitivity to H2O2-induced oxidative stress in vitro. Therefore, these results suggest that early life stress can have long-term effects on retinal structure and function, possibly elicited by neonatal immune preconditioning.
Collapse
Affiliation(s)
- Mariana Grigoruta
- Department of Chemical and Biological Sciences. Biomedical Sciences Institute. Autonomous University of Ciudad Juarez, Anillo envolvente Pronaf y Estocolmo S/N, Zona Pronaf, 32315 Ciudad Juárez, Chihuahua, Mexico; Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA
| | - Marbella Chavez-Solano
- Department of Chemical and Biological Sciences. Biomedical Sciences Institute. Autonomous University of Ciudad Juarez, Anillo envolvente Pronaf y Estocolmo S/N, Zona Pronaf, 32315 Ciudad Juárez, Chihuahua, Mexico; Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA.
| | - Armando Varela-Ramirez
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA
| | - Jorge A Sierra-Fonseca
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA
| | - Ernesto Orozco-Lucero
- Department of Veterinary Sciences. Biomedical Sciences Institute. Autonomous University of Ciudad Juarez, Anillo envolvente Pronaf y Estocolmo S/N, Zona Pronaf, 32315 Ciudad Juarez, Chihuahua, Mexico
| | - Jameel N Hamdan
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA
| | - Kristin L Gosselink
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, 500 West University Avenue, 79968 El Paso, TX, USA.
| | - Alejandro Martinez-Martinez
- Department of Chemical and Biological Sciences. Biomedical Sciences Institute. Autonomous University of Ciudad Juarez, Anillo envolvente Pronaf y Estocolmo S/N, Zona Pronaf, 32315 Ciudad Juárez, Chihuahua, Mexico
| |
Collapse
|
34
|
Abstract
This report describes the protocol for an ongoing project funded by the National Institutes of Health (R01MH108155) that is focused on effects of childhood maltreatment (MALTX) on neurocircuitry changes associated with adolescent major depressive disorder (MDD). Extant clinical and neuroimaging literature on MDD is reviewed, which has relied on heterogeneous samples that do not parse out the unique contribution of MALTX on neurobiological changes in MDD. Employing a 2 × 2 study design (controls with no MALTX or MDD, MALTX only, MDD only, and MDD + MALTX), and based on a cohesive theoretical model that incorporates behavioral, cognitive and neurobiological domains, we describe the multi-modal neuroimaging techniques used to test whether structural and functional alterations in the fronto-limbic and fronto-striatal circuits associated with adolescent MDD are moderated by MALTX. We hypothesize that MDD + MALTX youth will show alterations in the fronto-limbic circuit, with reduced connectivity between the amygdala (AMG) and the prefrontal cortex (PFC), as the AMG is sensitive to stress/threat during development. Participants with MDD will exhibit increased functional connectivity between the AMG and PFC due to self-referential negative emotions. Lastly, MDD + MALTX will only show changes in motivational/anticipatory aspects of the fronto-striatal circuit, and MDD will exhibit changes in motivational and consummatory/outcome aspects of reward-processing. Our goal is to identify distinct neural substrates associated with MDD due to MALTX compared to other causes, as these markers could be used to more effectively predict treatment outcome, index treatment response, and facilitate alternative treatments for adolescents who do not respond well to traditional approaches.
Collapse
|
35
|
Joss D, Lazar SW, Teicher MH. Effects of a mindfulness based behavioral intervention for young adults with childhood maltreatment history on hippocampal morphometry: a pilot MRI study with voxel-based morphometry. Psychiatry Res Neuroimaging 2020; 301:111087. [PMID: 32413812 PMCID: PMC7395365 DOI: 10.1016/j.pscychresns.2020.111087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/28/2020] [Accepted: 04/16/2020] [Indexed: 01/15/2023]
Abstract
Childhood maltreatment has long lasting impacts on neural development of the hippocampus, which is important for learning and memory. The present study aimed to assess the effects of a mindfulness based intervention on hippocampal morphometry and episodic memory in this population. We administered MRI, psychological questionnaires and an episodic memory task to 21 participants (5 males) before and after a mindfulness-based behavioral intervention, compared to 21 participants (7 males) on the waiting list. Changes in Gray Matter Volume (GMV) in bilateral hippocampi were analyzed with Voxel-Based Morphometry (VBM). One cluster was identified in the right hippocampus with a group by time interaction effect that consisted of 130 contiguous voxels but fell short of significance with full FDR correction (p = 0.077). GMV in this cluster increased by 0.76% in the mindfulness group and decreased by 0.78% in the control group. Within the mindfulness group, changes in hippocampal GMV were negatively associated with changes in perceived stress and depression severity and positively associated with enhancement in performance accuracy on the episodic memory task. Findings from this pilot study suggest that a mindfulness-based intervention may lead to an increase in partial hippocampal GMV with associated symptom reduction and improvement in episodic memory.
Collapse
Affiliation(s)
- Diane Joss
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Massachusetts General Hospital, USA; Department of Psychiatry, Harvard Medical School, Boston, USA.
| | - Sara W Lazar
- Department of Psychiatry, Massachusetts General Hospital, USA; Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Martin H Teicher
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, USA
| |
Collapse
|
36
|
Tata DA, Dandi E, Spandou E. Expression of synaptophysin and BDNF in the medial prefrontal cortex following early life stress and neonatal hypoxia-ischemia. Dev Psychobiol 2020; 63:173-182. [PMID: 32623722 DOI: 10.1002/dev.22011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 05/10/2020] [Accepted: 06/04/2020] [Indexed: 01/06/2023]
Abstract
This study aims at investigating whether early stress interacts with brain injury due to neonatal hypoxia-ischemia (HI). To this end, we examined possible changes in synaptophysin (SYN) and brain-derived neurotrophic factor (BDNF) expression in the medial prefrontal cortex (mPFC) of maternally separated rats that were subsequently exposed to a HI episode. Rat pups (n = 11) were maternally separated during postnatal days 1 to 6 (3hr/day), while another group was left undisturbed (n = 11). On postnatal day 7, a subgroup (n = 12) from each postnatal manipulation was exposed to HI. Synaptophysin and BDNF expression was estimated in mPFC prelimbic and anterior cingulate subregions of the ipsilateral and contralateral to the occluded common carotid artery hemispheres. Maternally separated rats expressed significantly less BDNF and SYN in both hemispheres. Neonatal HI significantly reduced BDNF and SYN expression in the ipsilateral mPFC only and this reduction was not further altered by early stress. Our findings indicate the enduring negative effect of a short period of maternal separation on the expression of mPFC SYN and BDNF. They, also, reveal that the HI-associated decreases in these markers are limited to the ipsilateral mPFC and are not exacerbated by early stress. These decreases may have important functional implications given the role of prefrontal area in high-order cognition.
Collapse
Affiliation(s)
- Despina A Tata
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evgenia Dandi
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Spandou
- Laboratory of Experimental Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
37
|
Yuan M, Rubin-Falcone H, Lin X, Rizk MM, Miller JM, Sublette ME, Oquendo MA, Burke A, Ogden RT, Mann JJ. Smaller left hippocampal subfield CA1 volume is associated with reported childhood physical and/or sexual abuse in major depression: A pilot study. J Affect Disord 2020; 272:348-354. [PMID: 32553377 DOI: 10.1016/j.jad.2020.03.169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/22/2020] [Accepted: 03/29/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Smaller hippocampal volumes are reported in adults with major depressive disorder (MDD) and in reported childhood abuse. The hippocampus is a complex structure with distinct functional subfields. We sought to examine the effect of MDD diagnosis and childhood abuse on hippocampal subfields. METHODS Forty-one MDD participants (17 reported abuse and 24 did not) and 46 healthy volunteers (HV) (2 reported abuse) underwent T1- weighted structural magnetic resonance imaging (MRI) and clinical characterization in a retrospective design. A subfield segmentation program was used to measure the whole and subfield hippocampal volumes. Linear mixed-effects models were fitted for group comparisons. RESULTS No main effect of diagnosis interaction effect between diagnosis and subfield region was observed. However, a comparison of abused MDD vs. HVs showed a group by region interaction. A significant interaction between childhood abuse and region was observed. Effects were confined to the left side of the brain, and post hoc, exploratory region-specific tests indicated smaller left CA1 volume in abused MDD compared with non-abused MDD. In addition, smaller amygdala volume was found in all MDD compared with HVs. LIMITATIONS We did not have a sample of healthy volunteers with reported childhood abuse. CONCLUSIONS The diagnosis of pure MDD may not be sufficient to exert effects on hippocampal volumes, indicating the importance of taking into account childhood trauma in studies on psychopathological mechanisms. Left CA1 might be the hippocampal subfield most relevant to reported childhood abuse. Smaller amygdala volume may be related to MDD diagnosis independent of childhood abuse.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China; Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States.
| | - Harry Rubin-Falcone
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Xuejing Lin
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Mina M Rizk
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States; Department of Psychiatry, Faculty of Medicine, Minia University, Egypt
| | - Jeffrey M Miller
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - M Elizabeth Sublette
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ainsley Burke
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - R Todd Ogden
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States; Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States; Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, United States.
| |
Collapse
|
38
|
Badura-Brack AS, Mills MS, Embury CM, Khanna MM, Klanecky Earl A, Stephen JM, Wang YP, Calhoun VD, Wilson TW. Hippocampal and parahippocampal volumes vary by sex and traumatic life events in children. J Psychiatry Neurosci 2020; 45:288-297. [PMID: 32078279 PMCID: PMC7828931 DOI: 10.1503/jpn.190013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Childhood trauma is reliably associated with smaller hippocampal volume in adults; however, this finding has not been shown in children, and even less is known about how sex and trauma interact to affect limbic structural development in children. METHODS Typically developing children aged 9 to 15 years who completed a trauma history questionnaire and structural T1-weighted MRI were included in this study (n = 172; 85 female, 87 male). All children who reported 4 or more traumas (n = 36) composed the high trauma group, and all children who reported 3 or fewer traumas (n = 136) composed the low trauma group. Using multivariate analysis of covariance, we compared FreeSurfer-derived structural MRI volumes (normalized by total intracranial volume) of the amygdalar, hippocampal and parahippocampal regions by sex and trauma level, controlling for age and study site. RESULTS We found a significant sex × trauma interaction, such that girls with high trauma had greater volumes than boys with high trauma. Follow-up analyses indicated significantly increased volumes for girls and generally decreased volumes for boys, specifically in the hippocampal and parahippocampalregions for the high trauma group; we observed no sex differences in the low trauma group. We noted no interaction effect for the amygdalae. LIMITATIONS We assessed a community sample and did not include a clinical sample. We did not collect data about the ages at which children experienced trauma. CONCLUSION Results revealed that psychological trauma affects brain development differently in girls and boys. These findings need to be followed longitudinally to elucidate how structural differences progress and contribute to well-known sex disparities in psychopathology.
Collapse
Affiliation(s)
- Amy S. Badura-Brack
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Mackenzie S. Mills
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Christine M. Embury
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Maya M. Khanna
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Alicia Klanecky Earl
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Julia M. Stephen
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Yu-Ping Wang
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Vince D. Calhoun
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| | - Tony W. Wilson
- From the Department of Psychological Science, Creighton University, Omaha, NE (Badura-Brack, Mills, Khanna, Klanecky Earl); the Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE (Embury, Wilson); the Department of Psychology, University of Nebraska at Omaha, Omaha, NE (Embury); the Mind Research Network, Albuquerque, NM (Stephen, Calhoun); and the Department of Biomedical Engineering, Tulane University, New Orleans, LA (Wang)
| |
Collapse
|
39
|
Region-specific effects of maternal separation on oxidative stress accumulation in parvalbumin neurons of male and female rats. Behav Brain Res 2020; 388:112658. [PMID: 32339550 DOI: 10.1016/j.bbr.2020.112658] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/16/2022]
Abstract
Early life adversity in humans is linked to cognitive deficits and increased risk of mental illnesses, including depression, bipolar disorder, and schizophrenia, with evidence for different vulnerabilities in men versus women. Modeling early life adversity in rodents shows similar neuropsychological deficits that may partially be driven by sex-dependent dysfunction in parvalbumin (PV) interneurons in the prefrontal cortex (PFC), hippocampus (HPC), and basolateral amygdala (BLA). Research demonstrates that PV interneurons are particularly susceptible to oxidative stress; therefore, accumulation of oxidative damage may drive PV dysfunction following early life adversity. The goal of this study was to quantify oxidative stress accumulation in PV neurons in rats exposed to maternal separation (MS). Pups were separated from their dam and littermates for 4 h per day from postnatal day (P)2 to 20. Serial sections from the PFC, HPC, and BLA of juvenile (P20) rats of both sexes were immunohistochemically stained with antibodies against PV and 8-oxo-dG, a marker for oxidative DNA damage. PV cell counts, colocalization with 8-oxo-dG, and intensity of each signal were measured in each region to determine the effects of MS and establish whether MS-induced oxidative damage varies between sexes. A significant increase in colocalization of PV and 8-oxo-dG was found in the PFC and HPC, indicating increased oxidative stress in that cell population following MS. Region-specific sex differences were also revealed in the PFC, BLA, and HPC. These data identify oxidative stress during juvenility as a potential mechanism mediating PV dysfunction in individuals with a history of early life adversity.
Collapse
|
40
|
Abstract
AbstractThe concept of minimal emotional dysfunctions (MED) refers to traditional psychopathology in order to describe, classify, and understand personality disorders. Emotional dysfunctions encompass disorders of affect predominance, production, expression, experience, modulation, and regulation. MED can explain the dimensional nature of personality disorders, their multidimensionality and problems with categorical classifications. It can stimulate research on the etiology of personality disorders in reference to modern developmental brain research and trauma psychology. It can guide new developments in pharmacotherapy and psychotherapy. It is suggested to focus on MED in future developments of the description and classification of personality disorders.
Collapse
Affiliation(s)
- M Linden
- Research Group Psychosomatic Rehabilitation at the Charité, University Medicine Berlin and the Department of Behavioral Medicine at the Rehabilitation Center Seehof, Lichterfelder Allee 55, Teltow 14513, Berlin, Germany.
| |
Collapse
|
41
|
Chan JC, Morgan CP, Adrian Leu N, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Neill Epperson C, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun 2020; 11:1499. [PMID: 32198406 PMCID: PMC7083921 DOI: 10.1038/s41467-020-15305-w] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 02/27/2020] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of incredible specificity in transmitting signals involved in cellular function, including germ cell maturation. Spermatogenesis occurs in the testes, behind a protective barrier to ensure safeguarding of germline DNA from environmental insults. Following DNA compaction, further sperm maturation occurs in the epididymis. Here, we report reproductive tract EVs transmit information regarding stress in the paternal environment to sperm, potentially altering fetal development. Using intracytoplasmic sperm injection, we found that sperm incubated with EVs collected from stress-treated epididymal epithelial cells produced offspring with altered neurodevelopment and adult stress reactivity. Proteomic and transcriptomic assessment of these EVs showed dramatic changes in protein and miRNA content long after stress treatment had ended, supporting a lasting programmatic change in response to chronic stress. Thus, EVs as a normal process in sperm maturation, can also perform roles in intergenerational transmission of paternal environmental experience.
Collapse
Affiliation(s)
- Jennifer C Chan
- Department of Biomedical Sciences, School of Veterinary Medicine and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christopher P Morgan
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - N Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amol Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yasmine M Cisse
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bridget M Nugent
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kathleen E Morrison
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Eldin Jašarević
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Weiliang Huang
- Department of Pharmaceutical Science, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Nickole Kanyuch
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ali B Rodgers
- Department of Biomedical Sciences, School of Veterinary Medicine and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Natarajan V Bhanu
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dara S Berger
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Benjamin A Garcia
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Seth Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Maureen Kane
- Department of Pharmaceutical Science, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - C Neill Epperson
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tracy L Bale
- Department of Pharmacology and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
42
|
Cotella EM, Morano RL, Wulsin AC, Martelle SM, Lemen P, Fitzgerald M, Packard BA, Moloney RD, Herman JP. Lasting Impact of Chronic Adolescent Stress and Glucocorticoid Receptor Selective Modulation in Male and Female Rats. Psychoneuroendocrinology 2020; 112:104490. [PMID: 31786480 PMCID: PMC7391799 DOI: 10.1016/j.psyneuen.2019.104490] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022]
Abstract
Adolescent animals are vulnerable to the effects of stress on brain development. We hypothesized that long-term effects of adolescent chronic stress are mediated by glucocorticoid receptor (GR) signaling. We used a specific GR modulator (CORT108297) to pharmacologically disrupt GR signaling in adolescent rats during exposure to chronic variable stress (CVS). Male and female rats received 30 mg/kg of drug during a 2-week CVS protocol starting at PND46. Emotional reactivity (open field) and coping behaviors (forced swim test (FST)) were then tested in adulthood, 5 weeks after the end of the CVS protocol. Blood samples were collected two days before FST and serial samples after the onset of the swim test to determine baseline and stress response levels of HPA hormones respectively. Our results support differential behavioral, physiological and stress circuit reactivity to adolescent chronic stress exposure in males and females, with variable involvement of GR signaling. In response to adolescent stress, males had heightened reactivity to novelty and exhibited marked reduction in neuronal excitation following swim stress in adulthood, whereas females developed a passive coping strategy in the FST and enhanced HPA axis stress reactivity. Only the latter effect was attenuated by treatment with the GR modulator C108297. In summary, our data suggest that adolescent stress differentially affects emotional behavior and circuit development in males and females, and that GR manipulation during stress can reverse at least some of these effects.
Collapse
MESH Headings
- Adaptation, Psychological/drug effects
- Adaptation, Psychological/physiology
- Age Factors
- Animals
- Aza Compounds/administration & dosage
- Aza Compounds/pharmacology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Female
- Heterocyclic Compounds, 4 or More Rings/administration & dosage
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Hypothalamo-Hypophyseal System/metabolism
- Hypothalamo-Hypophyseal System/physiopathology
- Male
- Rats
- Rats, Sprague-Dawley
- Receptors, Glucocorticoid/drug effects
- Receptors, Glucocorticoid/physiology
- Sex Factors
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Evelin M Cotella
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Rachel L Morano
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Aynara C Wulsin
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Susan M Martelle
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Paige Lemen
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Maureen Fitzgerald
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Benjamin A Packard
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Rachel D Moloney
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - James P Herman
- Dept. Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
43
|
María-Ríos CE, Morrow JD. Mechanisms of Shared Vulnerability to Post-traumatic Stress Disorder and Substance Use Disorders. Front Behav Neurosci 2020; 14:6. [PMID: 32082127 PMCID: PMC7006033 DOI: 10.3389/fnbeh.2020.00006] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Psychoactive substance use is a nearly universal human behavior, but a significant minority of people who use addictive substances will go on to develop an addictive disorder. Similarly, though ~90% of people experience traumatic events in their lifetime, only ~10% ever develop post-traumatic stress disorder (PTSD). Substance use disorders (SUD) and PTSD are highly comorbid, occurring in the same individual far more often than would be predicted by chance given the respective prevalence of each disorder. Some possible reasons that have been proposed for the relationship between PTSD and SUD are self-medication of anxiety with drugs or alcohol, increased exposure to traumatic events due to activities involved in acquiring illegal substances, or addictive substances altering the brain's stress response systems to make users more vulnerable to PTSD. Yet another possibility is that some people have an intrinsic vulnerability that predisposes them to both PTSD and SUD. In this review, we integrate clinical and animal data to explore these possible etiological links between SUD and PTSD, with an emphasis on interactions between dopaminergic, adrenocorticotropic, GABAergic, and glutamatergic neurobehavioral mechanisms that underlie different emotional learning styles.
Collapse
Affiliation(s)
| | - Jonathan D. Morrow
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
44
|
McQuaid GA, Darcey VL, Avalos MF, Fishbein DH, VanMeter JW. Altered cortical structure and psychiatric symptom risk in adolescents exposed to maternal stress in utero: A retrospective investigation. Behav Brain Res 2019; 375:112145. [PMID: 31400378 PMCID: PMC10561894 DOI: 10.1016/j.bbr.2019.112145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Maternal exposure to stress during pregnancy is associated with increased risk for cognitive and behavioral sequelae in offspring. Animal research demonstrates exposure to stress during gestation has effects on brain structure. In humans, however, little is known about the enduring effects of in utero exposure to maternal stress on brain morphology. We examine whether maternal report of stressful events during pregnancy is associated with brain structure and behavior in adolescents. We compare gray matter morphometry of typically-developing early adolescents (11-14 years of age, mean 12.7) at a single timepoint, based on presence/absence of retrospectively-assessed maternal report of negative major life event stress (MLES) during pregnancy: prenatal stress (PS; n = 28), comparison group (CG; n = 55). The Drug Use Screening Inventory Revised (DUSI-R) assessed adolescent risk for problematic behaviors. Exclusionary criteria included pre-term birth, low birth weight, and maternal substance use during pregnancy. Groups were equivalent for demographic (age, sex, IQ, SES, race/ethnicity), and birth measures (weight, length). Compared to CG peers, adolescents in the PS group exhibited increased gray matter density in bilateral posterior parietal cortex (PPC): bilateral intraparietal sulcus, left superior parietal lobule and inferior parietal lobule. Additionally, the PS group displayed greater risk for psychiatric symptoms and family system dysfunction, as assessed via DUSI-R subscales. These preliminary findings suggest that prenatal exposure to maternal MLES may exact enduring associations on offspring brain morphology and psychiatric risk, highlighting the importance of capturing these data in prospective longitudinal research studies (beginning at birth) to elucidate these associations.
Collapse
Affiliation(s)
- Goldie A McQuaid
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA.
| | - Valerie L Darcey
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA; Interdisciplinary Program in Neuroscience, Georgetown University, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| | - Melissa F Avalos
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| | - Diana H Fishbein
- Department of Human Development and Family Studies, Pennsylvania State University, 218 HHD Building, University Park, PA, 16802, USA
| | - John W VanMeter
- Center for Functional and Molecular Imaging, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC, 20057, USA
| |
Collapse
|
45
|
Music exposure attenuates anxiety- and depression-like behaviors and increases hippocampal spine density in male rats. Behav Brain Res 2019; 372:112023. [DOI: 10.1016/j.bbr.2019.112023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/18/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
|
46
|
Ulset VS, Czajkowski NO, Kraft B, Kraft P, Wikenius E, Kleppestø TH, Bekkhus M. Are unpopular children more likely to get sick? Longitudinal links between popularity and infectious diseases in early childhood. PLoS One 2019; 14:e0222222. [PMID: 31504058 PMCID: PMC6736236 DOI: 10.1371/journal.pone.0222222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/24/2019] [Indexed: 12/31/2022] Open
Abstract
Social stress and inflammatory processes are strong regulators of one another. Considerable evidence shows that social threats trigger inflammatory responses that increase infection susceptibility in both humans and animals, while infectious disease triggers inflammation that in turn regulates social behaviours. However, no previous study has examined whether young children’s popularity and their rate of infectious disease are associated. We investigated the longitudinal bidirectional links between children’s popularity status as perceived by peers, and parent reports of a variety of infectious diseases that are common in early childhood (i.e. common cold as well as eye, ear, throat, lung and gastric infections). We used data from the ‘Matter of the First Friendship Study’ (MOFF), a longitudinal prospective multi-informant study, following 579 Norwegian pre-schoolers (292 girls, median age at baseline = six years) with annual assessments over a period of three years. Social network analysis was used to estimate each child’s level of popularity. Cross-lagged autoregressive analyses revealed negative dose–response relations between children’s popularity scores and subsequent infection (b = –0.18, CI = –0.29, –0.06, and b = –0.13, CI = –0.23, –0.03). In conclusion, the results suggest that children who are unpopular in early childhood are at increased risk of contracting infection the following year.
Collapse
Affiliation(s)
| | | | - Brage Kraft
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Pål Kraft
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ellen Wikenius
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Mona Bekkhus
- Promenta Research Centre, Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Rakesh G, Morey RA, Zannas AS, Malik Z, Clausen A, Marx CE, Kritzer MD, Szabo ST. Resilience as a translational endpoint in the treatment of PTSD. Mol Psychiatry 2019; 24:1268-1283. [PMID: 30867558 PMCID: PMC6713904 DOI: 10.1038/s41380-019-0383-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 01/24/2019] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
Abstract
Resilience is a neurobiological entity that shapes an individual's response to trauma. Resilience has been implicated as the principal mediator in the development of mental illness following exposure to trauma. Although animal models have traditionally defined resilience as molecular and behavioral changes in stress responsive circuits following trauma, this concept needs to be further clarified for both research and clinical use. Here, we analyze the construct of resilience from a translational perspective and review optimal measurement methods and models. We also seek to distinguish between resilience, stress vulnerability, and posttraumatic growth. We propose that resilience can be quantified as a multifactorial determinant of physiological parameters, epigenetic modulators, and neurobiological candidate markers. This multifactorial definition can determine PTSD risk before and after trauma exposure. From this perspective, we propose the use of an 'R Factor' analogous to Spearman's g factor for intelligence to denote these multifactorial determinants. In addition, we also propose a novel concept called 'resilience reserve', analogous to Stern's cognitive reserve, to summarize the sum total of physiological processes that protect and compensate for the effect of trauma. We propose the development and application of challenge tasks to measure 'resilience reserve' and guide the assessment and monitoring of 'R Factor' as a biomarker for PTSD.
Collapse
Affiliation(s)
- Gopalkumar Rakesh
- Duke-UNC Brain Imaging and Analysis Center (BIAC), Durham, NC, 27710, USA. .,Durham VA Health Care System, Durham, NC, 27705, USA. .,VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center (MIRECC), 3022 Croasdaile Drive, Durham, NC, 27705, USA.
| | - Rajendra A Morey
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham NC, Duke University School of Medicine, Durham, NC 27710,VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center (MIRECC), 3022 Croasdaile Drive, Durham, NC 27705
| | | | - Zainab Malik
- Child and Adolescent Psychiatry, University of California, Davis, CA 95616
| | - Ashley Clausen
- Duke-UNC Brain Imaging and Analysis Center (BIAC), Durham VA Health Care System, VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center, 3022 Croasdaile Drive, Durham, NC 27705
| | - Christine E Marx
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA,Division of Translational Neurosciences, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Michael D Kritzer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Steven T Szabo
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA,Veterans Affairs Medical Center, Mental Health Service Line, Durham, North Carolina, 27710, USA
| |
Collapse
|
48
|
Matsudaira I, Oba K, Takeuchi H, Sekiguchi A, Tomita H, Taki Y, Kawashima R. rs1360780 of the FKBP5 gene modulates the association between maternal acceptance and regional gray matter volume in the thalamus in children and adolescents. PLoS One 2019; 14:e0221768. [PMID: 31465499 PMCID: PMC6715198 DOI: 10.1371/journal.pone.0221768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Investigating the effects of gene–environment interactions (G × E) with regard to brain structure may help to elucidate the putative mechanisms associated with psychiatric risk. rs1360780 (C/T) is a functional single-nucleotide polymorphism (SNP) in the gene encoding FK506–binding protein 5 (FKBP5), which is involved in the regulation of the hypothalamic–pituitary–adrenal (HPA) axis stress responses. The minor (T) allele of FKBP5 is considered a risk allele for stress-related disorders, due to the overproduction of FKBP5, which results in impaired communication of stress signals with the HPA axis. Previous studies have reported that interactions between childhood maltreatment and the rs1360780 genotype affect structures in subcortical areas of the brain. However, it is unclear how this SNP modulates the association between non-adverse environments and brain structure. In this study, we examined the interactive effect of the rs1360780 genotype and maternal acceptance on the regional gray matter volume (rGMV) in 202 Japanese children. Maternal acceptance was assessed using a Japanese psychological questionnaire for mothers. Whole-brain multiple regression analysis using voxel-based morphometry showed a significant positive association between maternal acceptance and rGMV in the left thalamus of T-allele carriers, while a significant negative association was found in C/C homozygotes. Post-hoc analysis revealed that at or below the 70th percentiles of maternal acceptance, the T-allele carriers had a reduced thalamic rGMV compared with that of C/C homozygotes. Thus, our investigation indicated that the effect of the maternal acceptance level on brain development was different, depending on the rs1360780 genotype. Importantly, we found that the differences in brain structure between the T-allele carriers and C/C homozygotes at low to moderate levels of maternal acceptance, which is not equivalent to maltreatment. The present study contributes to the G × E research by highlighting the necessity to investigate the role of non-adverse environmental factors.
Collapse
Affiliation(s)
- Izumi Matsudaira
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
- * E-mail:
| | - Kentaro Oba
- Department of Human Brain Science, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Hikaru Takeuchi
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Atsushi Sekiguchi
- Department of Behavioral Medicine, National Institute of Mental Health, National Center for Neurology and Psychiatry, Tokyo, Japan
- Division of Medical Neuroimaging Analysis, Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Psychiatry, Tohoku University Hospital, Sendai, Japan
| | - Yasuyuki Taki
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Division of Medical Neuroimaging Analysis, Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Smart-Aging Research Center, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Ryuta Kawashima
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Smart-Aging Research Center, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Department of Advanced Brain Science, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Smart Aging International Research Center, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
49
|
Doreste-Mendez R, Ríos-Ruiz EJ, Rivera-López LL, Gutierrez A, Torres-Reveron A. Effects of Environmental Enrichment in Maternally Separated Rats: Age and Sex-Specific Outcomes. Front Behav Neurosci 2019; 13:198. [PMID: 31555107 PMCID: PMC6727005 DOI: 10.3389/fnbeh.2019.00198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/13/2019] [Indexed: 01/10/2023] Open
Abstract
Maternal separation (MS) early in life is related to an increase in anxiety and depressive-like behaviors and neurobiological alterations mostly related to alterations in hypothalamic pituitary adrenal (HPA) axis reactivity. Environmental enrichment (EE) has been used to ameliorate the effects of MS. However, the outcomes of this intervention at different developmental periods after MS have not been studied. We subjected male and female Sprague–Dawley pups to MS and subsequently compared the effects of EE started either in the pre-pubertal period [postnatal day (PND) 22] or adulthood (PND 78). Anxiety and depressive-like behaviors as well as in hippocampal synaptic density and basal corticosterone, oxytocin, and vasopressin levels were measured. Our results support the beneficial effects of adulthood EE in decreasing anxiety in males as well as promoting synaptic density in ventral hippocampal CA3. Males displayed higher levels of vasopressin while females displayed higher oxytocin, with no changes in basal corticosterone for any group after EE.
Collapse
Affiliation(s)
- Raura Doreste-Mendez
- Department of Basic Sciences, Physiology and Pharmacology, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, United States.,School of Brain and Behavioral Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, United States
| | - Efraín J Ríos-Ruiz
- School of Brain and Behavioral Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, United States.,Institute of Translational Research in Behavioral Sciences, University of Puerto Rico-Ponce Campus, Ponce, PR, United States
| | - Leslie L Rivera-López
- Department of Neuroscience, University of Texas at Rio Grande Valley School of Medicine, Edinburg, TX, United States
| | - Alfredo Gutierrez
- Department of Community Health, School of Arts and Sciences, Tufts University, Medford, MA, United States
| | - Annelyn Torres-Reveron
- Department of Neuroscience, University of Texas at Rio Grande Valley School of Medicine, Edinburg, TX, United States.,Department of Human Genetics, University of Texas at Rio Grande Valley School of Medicine, Edinburg, TX, United States
| |
Collapse
|
50
|
The effects of sub-anesthetic ketamine plus ethanol on behaviors and apoptosis in the prefrontal cortex and hippocampus of adolescent rats. Pharmacol Biochem Behav 2019; 184:172742. [PMID: 31348944 DOI: 10.1016/j.pbb.2019.172742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/10/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
Abstract
Ketamine has become increasingly popular in adolescent drug abusers worldwide. Meanwhile, alcohol is usually used by ketamine users. However, little work has been conducted to examine the chronic combined effects of ketamine and ethanol on adolescent brain. Here we probed into the effects of chronic administration of ketamine at recreational doses alone or combined with ethanol on behaviors and neuron damage in an adolescent rat model. 28-day old rats were treated with either 20 or 30 mg/kg ketamine plus or not plus 10% ethanol daily for 21 days. Depressive like behaviors, anxiety like behavior and memory impairment were tested using open field test, forced swimming test, elevated plus maze and Morris water maze. Apoptosis in prefrontal cortex (PFC) and hippocampus (HIP) were determined by the TdT-mediated dUTP Nick-End Labeling (TUNEL) and protein and mRNA levels of caspase-3, Bax and Bcl-2. Results show that co-application of ketamine and ethanol significantly increased immobility time in the forced swimming test, up-regulated TUNEL positive cells and both protein and mRNA expressions of caspase-3 and Bax, compared with the control group and ketamine and ethanol use alone groups in the PFC, but not in the HIP. Our study suggests that chronic co-administration of ketamine and ethanol results in depressive-like behavior and the caspase-dependent apoptosis in the PFC of adolescent rats' brains.
Collapse
|