1
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
2
|
Sanguino-Gómez J, Huijgens S, den Hartog M, Schenk IJM, Kluck W, Versluis TD, Krugers HJ. Neural correlates of learning and memory are altered by early-life stress. Neurobiol Learn Mem 2024; 213:107952. [PMID: 38906243 DOI: 10.1016/j.nlm.2024.107952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
The ability to learn and remember, which is fundamental for behavioral adaptation, is susceptible to stressful experiences during the early postnatal period, such as abnormal levels of maternal care. The exact mechanisms underlying these effects still remain elusive. This study examined whether early life stress (ELS) alters memory and brain activation patterns in male mice. Therefore, we examined the expression of the immediate early genes (IEGs) c-Fos and Arc in the dentate gyrus (DG) and basolateral amygdala (BLA) after training and memory retrieval in a fear conditioning task. Furthermore, we examined the potential of RU38486 (RU486), a glucocorticoid receptor antagonist, to mitigate ELS-induced memory deficits by blocking stress signalling during adolescence. Arc::dVenus reporter mice, which allow investigating experience-dependent expression of the immediate early gene Arc also at more remote time points, were exposed to ELS by housing dams and offspring with limited bedding and nesting material (LBN) between postnatal days (PND) 2-9 and trained in a fear conditioning task at adult age. We found that ELS reduced both fear acquisition and contextual memory retrieval. RU486 did not prevent these effects. ELS reduced the number of Arc::dVenus+ cells in DG and BLA after training, while the number of c-Fos+ cells were left unaffected. After memory retrieval, ELS decreased c-Fos+ cells in the ventral DG and BLA. ELS also altered the colocalization of c-Fos+ cells with Arc::dVenus+ cells in the ventral DG, possibly indicating impaired engram allocation in the ventral DG after memory retrieval. In conclusion, this study shows that ELS alters neuronal activation patterns after fear acquisition and retrieval, which may provide mechanistic insights into enduring impact of ELS on the processing of fear memories, possibly via changes in cell (co-) activation and engram cell allocation.
Collapse
Affiliation(s)
| | - Stefan Huijgens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine den Hartog
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Inim J M Schenk
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Wenya Kluck
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara D Versluis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
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
|
4
|
Sanguino-Gómez J, Krugers HJ. Early-life stress impairs acquisition and retrieval of fear memories: sex-effects, corticosterone modulation, and partial prevention by targeting glucocorticoid receptors at adolescent age. Neurobiol Stress 2024; 31:100636. [PMID: 38883213 PMCID: PMC11177066 DOI: 10.1016/j.ynstr.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/11/2024] [Accepted: 04/20/2024] [Indexed: 06/18/2024] Open
Abstract
The early postnatal period is a sensitive time window that is characterized by several neurodevelopmental processes that define neuronal architecture and function later in life. Here, we examined in young adult mice, using an auditory fear conditioning paradigm, whether stress during the early postnatal period 1) impacts fear acquisition and memory consolidation in male and female mice; 2) alters the fear responsiveness to corticosterone and 3) whether effects of early-life stress (ELS) can be prevented by treating mice with a glucocorticoid (GR) antagonist at adolescence. Male and female mice were exposed to a limited nesting and bedding model of ELS from postnatal day (PND) 2-9 and injected i.p with RU38486 (RU486) at adolescent age (PND 28-30). At two months of age, mice were trained in the fear conditioning (FC) paradigm (with and without post training administration of corticosterone - CORT) and freezing behavior during fear acquisition and contextual and auditory memory retrieval was scored. We observed that ELS impaired fear acquisition specifically in male mice and reduced both contextual and auditory memory retrieval in male and female mice. Acute post-training administration of CORT increased freezing levels during auditory memory retrieval in female mice but reduced freezing levels during the tone presentation in particular in control males. Treatment with RU486 prevented ELS-effects in acquisition in male mice and in females during auditory memory retrieval. In conclusion, this study highlights the long-lasting consequences of early-life stress on fear memory processing and further illustrates 1) the potential of a glucocorticoid antagonist intervention during adolescence to mitigate these effects and 2) the partial modulation of the auditory retrieval upon post training administration of CORT, with all these effects being sex-dependent.
Collapse
Affiliation(s)
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Ferreira de Sá N, Camarini R, Suchecki D. One day away from mum has lifelong consequences on brain and behaviour. Neuroscience 2023:S0306-4522(23)00276-2. [PMID: 37352967 DOI: 10.1016/j.neuroscience.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
This chapter presents a brief overview of attachment theory and discusses the importance of the neonatal period in shaping an individual's physiological and behavioural responses to stress later in life, with a focus on the role of the parent-infant relationship, particularly in rodents. In rodents, the role of maternal behaviours goes far beyond nutrition, thermoregulation and excretion, acting as hidden regulators of the pup's physiology and development. In this review, we will discuss the inhibitory role of specific maternal behaviours on the ACTH and corticosterone (CORT) stress response. The interest of our group to explore the long-term consequences of maternal deprivation for 24 h (DEP) at different ages (3 days and 11 days) in rats was sparked by its opposite effects on ACTH and CORT levels. In early adulthood, DEP3 animals (males and females alike) show greater negative impact on affective behaviours and stress related parameters than DEP11, indicating that the latter is more resilient in tests of anxiety-like behaviour. These findings create an opportunity to explore the neurobiological underpinnings of vulnerability and resilience to stress-related disorders. The chapter also provides a brief historical overview and highlights the relevance of attachment theory, and how DEP helps to understand the effects of childhood parental loss as a risk factor for depression, schizophrenia, and PTSD in both childhood and adulthood. Furthermore, we present the concept of environmental enrichment (EE), its effects on stress responses and related behavioural changes and its benefits for rats previously subjected to DEP, along with the clinical implications of DEP and EE.
Collapse
Affiliation(s)
- Natália Ferreira de Sá
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo
| | - Rosana Camarini
- Department of Pharmacology - Instituto de Ciências Biomédicas, Universidade de São Paulo
| | - Deborah Suchecki
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo.
| |
Collapse
|
6
|
Huang Z, Jordan JD, Zhang Q. Early life adversity as a risk factor for cognitive impairment and Alzheimer's disease. Transl Neurodegener 2023; 12:25. [PMID: 37173751 PMCID: PMC10182702 DOI: 10.1186/s40035-023-00355-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Neurological conditions, including cognitive impairment and Alzheimer's disease (AD), impose a huge burden on society, affecting millions of people globally. In addition to genetic factors, recent studies indicate that environmental and experiential factors may contribute to the pathogenesis of these diseases. Early life adversity (ELA) has a profound impact on brain function and health later in life. In rodent models, exposure to ELA results in specific cognitive deficits and aggravated AD pathology. Extensive concerns have been raised regarding the higher risk of developing cognitive impairments in people with a history of ELA. In this review, we scrutinize findings from human and animal studies focusing on the connection of ELA with cognitive impairment and AD. These discoveries suggest that ELA, especially at early postnatal stages, increases susceptibility to cognitive impairment and AD later in life. In terms of mechanisms, ELA could lead to dysregulation of the hypothalamus-pituitary-adrenal axis, altered gut microbiome, persistent inflammation, oligodendrocyte dysfunction, hypomyelination, and aberrant adult hippocampal neurogenesis. Crosstalks among these events may synergistically contribute to cognitive impairment later in life. Additionally, we discuss several interventions that may alleviate adverse consequences of ELA. Further investigation into this crucial area will help improve ELA management and reduce the burden of related neurological conditions.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
7
|
Karst H, Droogers WJ, van der Weerd N, Damsteegt R, van Kroonenburg N, Sarabdjitsingh RA, Joëls M. Acceleration of GABA-switch after early life stress changes mouse prefrontal glutamatergic transmission. Neuropharmacology 2023; 234:109543. [PMID: 37061088 DOI: 10.1016/j.neuropharm.2023.109543] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 04/17/2023]
Abstract
Early life stress (ELS) alters the excitation-inhibition-balance (EI-balance) in various rodent brain areas and may be responsible for behavioral impairment later in life. The EI-balance is (amongst others) influenced by the switch of GABAergic transmission from excitatory to inhibitory, the so-called "GABA-switch". Here, we investigated how ELS affects the GABA-switch in mouse infralimbic Prefrontal Cortex layer 2/3 neurons, using the limited-nesting-and-bedding model. In ELS mice, the GABA-switch occurred already between postnatal day (P) 6 and P9, as opposed to P15-P21 in controls. This was associated with increased expression of the inward chloride transporter NKCC1, compared to the outward chloride transporter KCC2, both of which are important for the intracellular chloride concentration and, hence, the GABA reversal potential (Erev). Chloride transporters are not only important for regulating chloride concentration postsynaptically, but also presynaptically. Depending on the Erev of GABA, presynaptic GABAA receptor stimulation causes a depolarization or hyperpolarization, and thereby enhanced or reduced fusion of glutamate vesicles respectively, in turn changing the frequency of miniature postsynaptic currents (mEPSCs). In accordance, bumetanide, a blocker of NKCC1, shifted the Erev GABA towards more hyperpolarized levels in P9 control mice and reduced the mEPSC frequency. Other modulators of chloride transporters, e.g. VU0463271 (a KCC2 antagonist) and aldosterone -which increases NKCC1 expression-did not affect postsynaptic Erev in ELS P9 mice, but did increase the mEPSC frequency. We conclude that the mouse GABA-switch is accelerated after ELS, affecting both the pre- and postsynaptic chloride homeostasis, the former altering glutamatergic transmission. This may considerably affect brain development.
Collapse
Affiliation(s)
- Henk Karst
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Wouter J Droogers
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Nelleke van der Weerd
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Ruth Damsteegt
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Nicky van Kroonenburg
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - R Angela Sarabdjitsingh
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Marian Joëls
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
8
|
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: 7.0] [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
|
9
|
Sex-specific effects of neonatal paternal deprivation on microglial cell density in adult California mouse (Peromyscus californicus) dentate gyrus. Brain Behav Immun 2022; 106:1-10. [PMID: 35908654 DOI: 10.1016/j.bbi.2022.07.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Adverse early-life experiences are risk factors for psychiatric disease development, resulting in stress-related neuronal modeling and neurobehavioral changes. Stressful experiences modulate the immune system, contributing to neuronal damage in higher cortical regions, like the hippocampus. Moreover, early-life stressors dysregulate the function of microglia, the resident immune cells of the brain, in the developing hippocampus. Paternal deprivation, an early-life stressor in many biparental species, facilitates sex-dependent inhibitions in hippocampal plasticity, but parental contributors to these sex-specific outcomes are unknown. Also, neurobiological mechanisms contributing to impairments in hippocampal neuroplasticity are less known. Thus, our goals were to 1) determine whether parental behavior is altered in maternal females following removal of the paternal male, 2) assess the effects of paternal deprivation on dentate gyrus (DG) volume and microglia proliferation, and 3) determine if early-life experimental handling mitigates sex-specific reductions in DG cell survival. California mice were born to multiparous breeders and reared by both parents (biparental care) or by their mother alone (i.e., father removed on postnatal day 1; paternal deprivation). One cohort of offspring underwent offspring retrieval tests for eight days beginning on postnatal day 2. On PND 68, these offspring (and a second cohort of mice without behavioral testing) were euthanized and brains visualized for bromodeoxyuridine (BrdU) and neuron-specific class III beta-tubulin (TuJ-1) or ionized calcium binding adaptor molecule 1 (Iba1). While mate absence did not impair maternal retrieval, paternal deprivation reduced DG volume, but Iba1+ cell density was only higher in paternally-deprived females. Neither sex or paternal deprivation significantly altered the number of BrdU+ or Tuj1+ cells in the DG - an absence of a reduction in cell survival may be related to daily handing during early offspring retrieval tests. Together, these data suggest that paternal deprivation impairs hippocampal plasticity; however, sex and early environment may influence the magnitude of these outcomes.
Collapse
|
10
|
Garvin MM, Bolton JL. Sex-specific behavioral outcomes of early-life adversity and emerging microglia-dependent mechanisms. Front Behav Neurosci 2022; 16:1013865. [PMID: 36268470 PMCID: PMC9577368 DOI: 10.3389/fnbeh.2022.1013865] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Early-life adversity (ELA) is known to alter brain circuit maturation as well as increase vulnerability to cognitive and emotional disorders. However, the importance of examining sex as a biological variable when researching the effects of ELA has not been considered until recently. This perspective discusses the sex-specific behavioral outcomes of ELA in both humans and animal models, then proposes microglia-mediated mechanisms as a potential underlying cause. Recent work in rodent models suggests that ELA provokes cognitive deficits, anhedonia, and alcohol abuse primarily in males, whereas females exhibit greater risk-taking and opioid addiction-related behaviors. In addition, emerging evidence identifies microglia as a key target of ELA. For example, we have recently shown that ELA inhibits microglial synapse engulfment and process dynamics in male mice, leading to an increase in excitatory synapse number onto corticotrophin-releasing hormone (CRH)-expressing neurons in the paraventricular nucleus of the hypothalamus (PVN) and aberrant stress responses later in life. However, ELA-induced synaptic rewiring of neural circuits differs in females during development, resulting in divergent behavioral outcomes. Thus, examining the role of microglia in the sex-specific mechanisms underlying ELA-induced neuropsychiatric disorders is an important topic for future research.
Collapse
Affiliation(s)
| | - Jessica L. Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
11
|
Smith IF, Gursky ZH, Klintsova AY. Representation of prefrontal axonal efferents in the thalamic nucleus reuniens in a rodent model of fetal alcohol exposure during third trimester. Front Behav Neurosci 2022; 16:993601. [PMID: 36160686 PMCID: PMC9493097 DOI: 10.3389/fnbeh.2022.993601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol exposure (AE) during the prenatal period could result in fetal alcohol spectrum disorders (FASDs), one of many deficits of which is impaired executive functioning (EF). EF relies on the coordination of activity between the medial prefrontal cortex (mPFC) and hippocampus (HPC) by the thalamic nucleus reuniens (Re), a structure that has been shown to be damaged following high-dose AE in a rodent model of third trimester exposure. Notably, mPFC neurons do not project directly to HPC, but rather communicate with it via a disynaptic pathway where the first cortical axons synapse on neurons in Re, which in turn send axons to make contacts with hippocampal cells. This experiment investigated the effect of binge AE (5.25 g/kg/day, two doses 2 h apart) during postnatal days 4-9 on the length of medial prefrontal axonal projections within Re in Long Evans rat. AE reduced the cumulative length of mPFC-originating axon terminals in Re in female rats, with male rats exhibiting shorter cumulative lengths when compared to female procedural control animals. Additionally, Re volume was decreased in AE animals, a finding that reproduced previously reported data. This experiment helps us better understand how early life AE affects prefrontal-thalamic-hippocampal connectivity that could underlie subsequent EF deficits.
Collapse
Affiliation(s)
- Ian F. Smith
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE, United States
| | - Zachary H. Gursky
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Anna Y. Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
12
|
Barroca NCB, Della Santa G, Suchecki D, García-Cairasco N, Umeoka EHDL. Challenges in the use of animal models and perspectives for a translational view of stress and psychopathologies. Neurosci Biobehav Rev 2022; 140:104771. [PMID: 35817171 DOI: 10.1016/j.neubiorev.2022.104771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 12/25/2022]
Abstract
The neurobiology and development of treatments for stress-related neuropsychiatric disorders rely heavily on animal models. However, the complexity of these disorders makes it difficult to model them entirely, so only specific features of human psychopathology are emulated and these models should be used with great caution. Importantly, the effects of stress depend on multiple factors, like duration, context of exposure, and individual variability. Here we present a review on pre-clinical studies of stress-related disorders, especially those developed to model posttraumatic stress disorder, major depression, and anxiety. Animal models provide relevant evidence of the underpinnings of these disorders, as long as face, construct, and predictive validities are fulfilled. The translational challenges faced by scholars include reductionism and anthropomorphic/anthropocentric interpretation of the results instead of a more naturalistic and evolutionary understanding of animal behavior that must be overcome to offer a meaningful model. Other limitations are low statistical power of analysis, poor evaluation of individual variability, sex differences, and possible conflicting effects of stressors depending on specific windows in the lifespan.
Collapse
Affiliation(s)
- Nayara Cobra Barreiro Barroca
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Giovanna Della Santa
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Norberto García-Cairasco
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil; Department of Physiology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Eduardo Henrique de Lima Umeoka
- Department of Neuroscience and Behavioral Science, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil; School of Medicine, University Center UniCerrado, Goiatuba, GO, Brazil
| |
Collapse
|
13
|
Upadhyaya K, Sharma PK, Akhtar A, Pilkhwal Sah S. Protective Effects of Zingerone Against Depression-Like Behavior and Biochemical Changes in Chronic Stressed Rats: Antioxidant Effects. J Med Food 2022; 25:576-587. [PMID: 35639359 DOI: 10.1089/jmf.2021.k.0141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ginger contains zingerone, an active constituent possessing antioxidant and neuroprotective properties. The present study was designed to explore the efficacy of the bioactive compound, zingerone, for treating behavioral and biochemical alterations in rats exposed to chronic restraint stress (CRS). Female Wistar rats were administered zingerone (25, 50, and 100 mg/kg p.o.) once daily for a period of 28 days while being exposed to CRS (6 h/day). Our results indicated that the stressed animals depicted depression-like behavior (reduced sucrose preference and increased immobility time) associated with increased lipid peroxidation (LPO) (cortex), decreased catalase (CAT) (hippocampus and cortex), and increased superoxide dismutase (SOD) (hippocampus and cortex). In addition, metabolic alterations were characterized by hyperglycemia and increased glycosylated hemoglobin in the CRS rats. However, no alterations were observed for learning and memory and in the levels of reduced glutathione. Repeated zingerone administration significantly reversed depression-like behavior elicited by CRS in rats. Furthermore, a significant antioxidant effect was exhibited by zingerone, as shown by decreased LPO and enhanced activity of SOD and CAT in chronically stressed rats. The findings of our study demonstrated that zingerone possesses protective actions against chronic stress-induced depressive-like behavioral, biochemical, and metabolic alterations and that its underlying mechanism may be attributed to its antioxidant properties. The results also signify its pharmacological and possible nutritional importance.
Collapse
Affiliation(s)
- Kumud Upadhyaya
- Department of Pharmaceutical Sciences, Kumaun University, Nainital, India
| | | | - Ansab Akhtar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, India
| | - Sangeeta Pilkhwal Sah
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, India
| |
Collapse
|
14
|
Effects of early life adversities upon memory processes and cognition in rodent models. Neuroscience 2022; 497:282-307. [PMID: 35525496 DOI: 10.1016/j.neuroscience.2022.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/14/2023]
Abstract
Exposure to stressors in early postnatal life induces long-lasting modifications in brainfunction.Thisplasticity,an essential characteristic of the brain that enables adaptation to the environment, may also induce impairments in some psychophysiological functions, including learning and memory. Early life stress (ELS) has long-term effects on thehypothalamic-pituitary-adrenal axisresponse to stressors, and has been reported to lead toneuroinflammation,altered levelsof neurotrophic factors, modifications inneurogenesis andsynaptic plasticity,with changes in neurotransmitter systems and network functioning. In this review, we focus on early postnatal stress in animal models and their effects on learning and memory.Many studies have reported ELS-induced impairments in different types of memories, including spatial memory, fear memory, recognition (both for objects and social) memory, working memory and reversal learning. Studies are not always in agreement, however, no effects, or sometimes facilitation, being reported, depending on the nature and intensity of the early intervention, as well as the age when the outcome was evaluated and the sex of the animals. When considering processes occurring after consolidation, related with memory maintenance or modification, there are a very reduced number of reports. Future studies addressing the mechanisms underlying memory changes for ELS should shed some light on the understanding of the different effects induced by stressors of different types and intensities on cognitive functions.
Collapse
|
15
|
Roque A, Valles Méndez KM, Ruiz R, Pineda E, Lajud N. Early life stress induces a transient increase in hippocampal corticotropin-releasing hormone in rat neonates that precedes the effects on hypothalamic neuropeptides. Eur J Neurosci 2022; 55:2108-2121. [PMID: 33745155 DOI: 10.1111/ejn.15193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Early life stress (ELS) programs hypothalamus-pituitary-adrenal (HPA) axis activity and affects synaptic plasticity and cognitive performance in adults; however, the effects of ELS during the temporal window of vulnerability are poorly understood. This study aimed to thoroughly characterize the effects of ELS in the form of periodic maternal separation (MS180) during the time of exposure to stress. Hippocampal corticotropin-releasing hormone (CRH) gene expression and baseline HPA axis activity were analyzed at postnatal (P) days 6, 12, 15, and 21, and in adulthood (P75); these factors were correlated with plasticity markers and adult behavior. Our results indicate that MS180 induces an increase in hippocampal CRH expression at P9, P12, and P15, whereas an increase in hypothalamic CRH expression was observed from P12 to P21. Increased arginine-vasopressin expression and corticosterone levels were observed only at P21. Moreover, MS180 caused transient alterations in hypothalamic synaptophysin expression during early life. As adults, MS180 rats showed a passive coping strategy in the forced swimming test, cognitive impairments in the object location test, increased hypothalamic CRH expression, and decreased oxytocin (OXT) expression. Spearman's analysis indicated that cognitive impairments correlated with CRH and OXT expression. In conclusion, our data indicate that MS180 induces a transient increase in hippocampal CRH expression in neonates that precedes the effects on hypothalamic neuropeptides, confirming the role of increased CRH during the temporal window of vulnerability as a mediator of some of the detrimental effects of ELS on brain development and adult behavior.
Collapse
Affiliation(s)
- Angélica Roque
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Kinberli Marcela Valles Méndez
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Roberto Ruiz
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Edel Pineda
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Naima Lajud
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| |
Collapse
|
16
|
Colettis NC, Habif M, Oberholzer MV, Filippin F, Jerusalinsky DA. Differences in learning and memory between middle-aged female and male rats. Learn Mem 2022; 29:120-125. [PMID: 35428728 PMCID: PMC9053109 DOI: 10.1101/lm.053578.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
We observed differences in cognitive functions between middle-aged female and male Wistar rats. Both (like youngsters) discriminated new versus familiar objects, showing similar short- and long-term memory (STM and LTM, respectively). Only females show robust LTM for new location of an object. Both successfully form LTM of inhibitory avoidance, though males appeared to be amnesic for memory persistence. Habituation, locomotion, horizontal exploration, "stereotypies," fear, and anxiety-like behavior were similar for both, while vertical exploration was significantly higher in middle-aged and younger females. Therefore, sex-dependent differences in some cognitive functions and behaviors must be considered when designing and interpreting learning and memory studies.
Collapse
Affiliation(s)
- Natalia Claudia Colettis
- Laboratorio de Neuroplasticidad y Neurotoxinas (LaN&N), Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Medicina, Buenos Aires C1121ABG, Argentina.,Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica. Buenos Aires C1113AAD, Argentina
| | - Martín Habif
- Laboratorio de Neuroplasticidad y Neurotoxinas (LaN&N), Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - María Victoria Oberholzer
- Laboratorio de Neuroplasticidad y Neurotoxinas (LaN&N), Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Federico Filippin
- Laboratorio de Neuroplasticidad y Neurotoxinas (LaN&N), Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Diana Alicia Jerusalinsky
- Laboratorio de Neuroplasticidad y Neurotoxinas (LaN&N), Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
17
|
The Problem of Malnutrition Associated with Major Depressive Disorder from a Sex-Gender Perspective. Nutrients 2022; 14:nu14051107. [PMID: 35268082 PMCID: PMC8912662 DOI: 10.3390/nu14051107] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 02/03/2023] Open
Abstract
Major depressive disorder (MDD) is an incapacitating condition characterized by loss of interest, anhedonia and low mood, which affects almost 4% of people worldwide. With rising prevalence, it is considered a public health issue that affects economic productivity and heavily increases health costs alone or as a comorbidity for other pandemic non-communicable diseases (such as obesity, cardiovascular disease, diabetes, inflammatory bowel diseases, etc.). What is even more noteworthy is the double number of women suffering from MDD compared to men. In fact, this sex-related ratio has been contemplated since men and women have different sexual hormone oscillations, where women meet significant changes depending on the age range and moment of life (menstruation, premenstruation, pregnancy, postpartum, menopause…), which seem to be associated with susceptibility to depressive symptoms. For instance, a decreased estrogen level promotes decreased activation of serotonin transporters. Nevertheless, sexual hormones are not the only triggers that alter neurotransmission of monoamines and other neuropeptides. Actually, different dietary habits and/or nutritional requirements for specific moments of life severely affect MDD pathophysiology in women. In this context, the present review aims to descriptively collect information regarding the role of malnutrition in MDD onset and course, focusing on female patient and especially macro- and micronutrient deficiencies (amino acids, ω3 polyunsaturated fatty acids (ω3 PUFAs), folate, vitamin B12, vitamin D, minerals…), besides providing evidence for future nutritional intervention programs with a sex-gender perspective that hopefully improves mental health and quality of life in women.
Collapse
|
18
|
Surakul P, Chutabhakdikul N, Vanichviriyakit R, Promthep K, Thangnipon W. Maternal Stress Induced Autophagy Dysfunction and Immune Activation in the Hippocampus of Adolescence Rat Pups. J Chem Neuroanat 2022; 121:102085. [DOI: 10.1016/j.jchemneu.2022.102085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 11/24/2022]
|
19
|
Kim S, Gacek SA, Mocchi MM, Redei EE. Sex-Specific Behavioral Response to Early Adolescent Stress in the Genetically More Stress-Reactive Wistar Kyoto More Immobile, and Its Nearly Isogenic Wistar Kyoto Less Immobile Control Strain. Front Behav Neurosci 2022; 15:779036. [PMID: 34970127 PMCID: PMC8713037 DOI: 10.3389/fnbeh.2021.779036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic predisposition and environmental stress are known etiologies of stress-related psychiatric disorders. Environmental stress during adolescence is assumed to be particularly detrimental for adult affective behaviors. To investigate how genetic stress-reactivity differences modify the effects of stress during adolescence on adult affective behaviors we employed two inbred strains with differing stress reactivity. The Wistar Kyoto More Immobile (WMI) rat strain show increased stress-reactivity and despair-like behaviors as well as passive coping compared to the nearly isogenic control strain, the Wistar Kyoto Less Immobile (WLI). Males and females of these strains were exposed to contextual fear conditioning (CFC) during early adolescence (EA), between 32 and 34 postnatal days (PND), and were tested for the consequences of this mild EA stress in adulthood. Early adolescent stress significantly decreased anxiety-like behavior, measured in the open field test (OFT) and increased social interaction and recognition in adult males of both strains compared to controls. In contrast, no significant effects of EA stress were observed in adult females in these behaviors. Both males and females of the genetically less stress-reactive WLI strain showed significantly increased immobility in the forced swim test (FST) after EA stress compared to controls. In contrast, immobility was significantly attenuated by EA stress in adult WMI females compared to controls. Transcriptomic changes of the glucocorticoid receptor (Nr3c1, GR) and the brain-derived neurotrophic factor (Bdnf) illuminate primarily strain and stress-dependent changes, respectively, in the prefrontal cortex and hippocampus of adults. These results suggest that contrary to expectations, limited adolescent stress is beneficial to males thru decreasing anxiety and enhancing social behaviors, and to the stress more-reactive WMI females by way of decreasing passive coping.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Stephanie A Gacek
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Madaline M Mocchi
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
20
|
Kahnau P, Guenther A, Boon MN, Terzenbach JD, Hanitzsch E, Lewejohann L, Brust V. Lifetime Observation of Cognition and Physiological Parameters in Male Mice. Front Behav Neurosci 2021; 15:709775. [PMID: 34539359 PMCID: PMC8442583 DOI: 10.3389/fnbeh.2021.709775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
Laboratory mice are predominantly used for one experiment only, i.e., new mice are ordered or bred for every new experiment. Moreover, most experiments use relatively young mice in the range of late adolescence to early adulthood. As a consequence, little is known about the day-to-day life of adult and aged laboratory mice. Here we present a long-term data set with three consecutive phases conducted with the same male mice over their lifetime in order to shed light on possible long-term effects of repeated cognitive stimulation. One third of the animals was trained by a variety of learning tasks conducted up to an age of 606 days. The mice were housed in four cages with 12 animals per cage; only four mice per cage had to repeatedly solve cognitive tasks for getting access to water using the IntelliCage system. In addition, these learner mice were tested in standard cognitive tests outside their home-cage. The other eight mice served as two control groups living in the same environment but without having to solve tasks for getting access to water. One control group was additionally placed on the test set-ups without having to learn the tasks. Next to the cognitive tasks, we took physiological measures (body mass, resting metabolic rate) and tested for dominance behavior, and attractivity in a female choice experiment. Overall, the mice were under surveillance until they died a natural death, providing a unique data set over the course of virtually their entire lives. Our data showed treatment differences during the first phase of our lifetime data set. Young learner mice showed a higher activity, less growth and resting metabolic rate, and were less attractive for female mice. These effects, however, were not preserved over the long-term. We also did not find differences in dominance or effects on longevity. However, we generated a unique and valuable set of long-term behavioral and physiological data from a single group of male mice and note that our long-term data contribute to a better understanding of the behavioral and physiological processes in male C57Bl/6J mice.
Collapse
Affiliation(s)
- Pia Kahnau
- Laboratory Animal Science, German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Anja Guenther
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
| | - Marcus Nicolaas Boon
- Department for Electrical Engineering and Computer Science, Modeling of Cognitive Processes, Technische Universität Berlin, Berlin, Germany
- Exzellenzcluster Science of Intelligence, Technische Universität Berlin, Berlin, Germany
| | | | - Eric Hanitzsch
- Behavioral Phenotyping Unit, University of Osnabrück, Osnabrück, Germany
| | - Lars Lewejohann
- Laboratory Animal Science, German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment (BfR), Berlin, Germany
- Animal Behavior and Laboratory Animal Science, Institute of Animal Welfare, Freie Universität Berlin, Berlin, Germany
| | - Vera Brust
- Behavioral Phenotyping Unit, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
21
|
Sex-specific behavioral and structural alterations caused by early-life stress in C57BL/6 and BTBR mice. Behav Brain Res 2021; 414:113489. [PMID: 34303728 DOI: 10.1016/j.bbr.2021.113489] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/06/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022]
Abstract
Lately, the development of various mental illnesses, such as depression, personality disorders, and autism spectrum disorders, is often associated with traumatic events in childhood. Nonetheless, the mechanism giving rise to this predisposition is still unknown. Because the development of a disease often depends on a combination of a genetic background and environment, we decided to evaluate the effect of early-life stress on BTBR mice, which have behavioral, neuroanatomical, and physiological features of autism spectrum disorders. As early-life stress, we used prolonged separation of pups from their mothers in the first 2 weeks of life (3 h once a day). We assessed effects of the early-life stress on juvenile (postnatal day 23) and adolescent (postnatal days 37-38) male and female mice of strains C57BL/6 (B6) and BTBR. We found that in both strains, the early-life stress did not lead to changes in the level of social behavior, which is an important characteristic of autism-related behavior. Nonetheless, the early-life stress resulted in increased locomotor activity in juvenile BTBR mice. In adolescent mice, the stress early in life caused a low level of anxiety in B6 males and BTBR females and increased exploratory activity in adolescent BTBR males and females. In addition, adolescent B6 male and female mice with a history of the early-life stress tended to have a thinner motor cortex as assessed by magnetic resonance imaging. As compared to B6 mice, BTBR mice showed reduced levels of social behavior and exploratory activity but their level of locomotor activity was higher. BTBR mice had smaller whole-brain, cortical, and dorsal hippocampal volumes; decreased motor cortex thickness; and increased ventral-hippocampus volume as compared to B6 mice, and these parameters correlated with the level of exploratory behavior of BTBR mice. Overall, the effects of early postnatal stress are sex- and strain-dependent.
Collapse
|
22
|
Eachus H, Choi MK, Ryu S. The Effects of Early Life Stress on the Brain and Behaviour: Insights From Zebrafish Models. Front Cell Dev Biol 2021; 9:657591. [PMID: 34368117 PMCID: PMC8335398 DOI: 10.3389/fcell.2021.657591] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/20/2021] [Indexed: 01/27/2023] Open
Abstract
The early life period represents a window of increased vulnerability to stress, during which exposure can lead to long-lasting effects on brain structure and function. This stress-induced developmental programming may contribute to the behavioural changes observed in mental illness. In recent decades, rodent studies have significantly advanced our understanding of how early life stress (ELS) affects brain development and behaviour. These studies reveal that ELS has long-term consequences on the brain such as impairment of adult hippocampal neurogenesis, altering learning and memory. Despite such advances, several key questions remain inadequately answered, including a comprehensive overview of brain regions and molecular pathways that are altered by ELS and how ELS-induced molecular changes ultimately lead to behavioural changes in adulthood. The zebrafish represents a novel ELS model, with the potential to contribute to answering some of these questions. The zebrafish offers some important advantages such as the ability to non-invasively modulate stress hormone levels in a whole animal and to visualise whole brain activity in freely behaving animals. This review discusses the current status of the zebrafish ELS field and its potential as a new ELS model.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Min-Kyeung Choi
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Soojin Ryu
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom.,Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
23
|
Ceci FM, Ferraguti G, Petrella C, Greco A, Tirassa P, Iannitelli A, Ralli M, Vitali M, Ceccanti M, Chaldakov GN, Versacci P, Fiore M. Nerve Growth Factor, Stress and Diseases. Curr Med Chem 2021; 28:2943-2959. [PMID: 32811396 DOI: 10.2174/0929867327999200818111654] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 11/22/2022]
Abstract
Stress is a constant threat for homeostasis and is represented by different extrinsic and intrinsic stimuli (stressors, Hans Selye's "noxious agents"), such as aggressive behavior, fear, diseases, physical activity, drugs, surgical injury, and environmental and physiological changes. Our organisms respond to stress by activating the adaptive stress system to activate compensatory responses for restoring homeostasis. Nerve Growth Factor (NGF) was discovered as a signaling molecule involved in survival, protection, differentiation, and proliferation of sympathetic and peripheral sensory neurons. NGF mediates stress with an important role in translating environmental stimuli into physiological and pathological feedbacks since NGF levels undergo important variations after exposure to stressful events. Psychological stress, lifestyle stress, and oxidative stress are well known to increase the risk of mental disorders such as schizophrenia, major depressive disorders, bipolar disorder, alcohol use disorders and metabolic disorders such as metabolic syndrome. This review reports recent works describing the activity of NGF in mental and metabolic disorders related to stress.
Collapse
Affiliation(s)
- Flavio Maria Ceci
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Angela Iannitelli
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | | | - Mauro Ceccanti
- Centro Riferimento Alcologico Regione Lazio, ASL Roma 1, Rome, Italy
| | - George N Chaldakov
- Department of Anatomy and Cell Biology, Medical University, and Institute for Advanced Study, Varna, Bulgaria
| | - Paolo Versacci
- Department of Pediatrics, Sapienza University Hospital of Rome, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| |
Collapse
|
24
|
Deficits in hippocampal-dependent memory across different rodent models of early life stress: systematic review and meta-analysis. Transl Psychiatry 2021; 11:231. [PMID: 33879774 PMCID: PMC8058062 DOI: 10.1038/s41398-021-01352-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/20/2021] [Accepted: 04/01/2021] [Indexed: 02/02/2023] Open
Abstract
Exposure to early life stress (ELS) causes abnormal hippocampal development and functional deficits in rodents and humans, but no meta-analysis has been used yet to quantify the effects of different rodent models of ELS on hippocampal-dependent memory. We searched PubMed and Web of Science for publications that assessed the effects of handling, maternal separation (MS), and limited bedding and nesting (LBN) on performance in the Morris water maze (MWM), novel object recognition (NOR), and contextual fear conditioning (CFC). Forty-five studies met inclusion criteria (n = 451-763 rodents per test) and were used to calculate standardized mean differences (Hedge's g) and to assess heterogeneity, publication bias, and the moderating effects of sex and species (rats vs. mice). We found significantly lower heterogeneity in LBN compared to handling and MS with no consistent effects of sex or species across the three paradigms. LBN and MS caused similar cognitive deficits in tasks that rely heavily on the dorsal hippocampus, such as MWM and NOR, and were significantly different compared to the improved performance seen in rodents exposed to handling. In the CFC task, which relies more on the ventral hippocampus, all three paradigms showed reduced freezing with moderate effect sizes that were not statistically different. These findings demonstrate the utility of using meta-analysis to quantify outcomes in a large number of inconsistent preclinical studies and highlight the need to further investigate the possibility that handling causes different alterations in the dorsal hippocampus but similar outcomes in the ventral hippocampus when compared to MS and LBN.
Collapse
|
25
|
Liu X, Sun YX, Zhang CC, Zhang XQ, Zhang Y, Wang T, Ma YN, Wang H, Su YA, Li JT, Si TM. Vortioxetine attenuates the effects of early-life stress on depression-like behaviors and monoamine transporters in female mice. Neuropharmacology 2021; 186:108468. [PMID: 33485943 DOI: 10.1016/j.neuropharm.2021.108468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/23/2020] [Accepted: 01/17/2021] [Indexed: 01/06/2023]
Abstract
Major depressive disorder is a major psychiatric disorder and a leading cause of disability around the world. Females have about twice as high an incidence of depression as males. However, preclinical animal models of depression have seldom investigated the molecular alterations associated with higher depression risk in females. In this study, adopting the early-life stress (ELS) paradigm of limited bedding and nesting material, we found that ELS induced depression-like behaviors only in adult female mice, as evaluated by sucrose preference and tail suspension tests. We then examined the ELS effects on monoamine neurotransmission (transporters for monoamine reuptake and release) in depression-related brain regions in female mice. We found that ELS resulted in widespread changes of the expression levels of these transporters in four brain regions. Moreover, systemic 21-day treatment with vortioxetine, a novel multimodal antidepressant, successfully reversed depression-like behaviors and normalized some molecular changes, including that of the norepinephrine transporter in the medial prefrontal cortex, vesicular monoamine transporter 2 in nucleus accumbens core, and serotonin transporter in amygdala. Collectively, these results provide evidence for the validity of using the limited bedding and nesting material paradigm to investigate sex differences in depression and demonstrate that the region-specific alterations of monoamine neurotransmission may be associated with depression-like behaviors in female mice. This article is part of the special issue on 'Stress, Addiction and Plasticity'.
Collapse
Affiliation(s)
- Xiao Liu
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Ya-Xin Sun
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Chen-Chen Zhang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Xian-Qiang Zhang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Yue Zhang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Ting Wang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Yu-Nu Ma
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Han Wang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Yun-Ai Su
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Ji-Tao Li
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China.
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, The Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China.
| |
Collapse
|
26
|
Oldham Green N, Maniam J, Riese J, Morris MJ, Voineagu I. Transcriptomic signature of early life stress in male rat prefrontal cortex. Neurobiol Stress 2021; 14:100316. [PMID: 33796639 PMCID: PMC7995657 DOI: 10.1016/j.ynstr.2021.100316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
Early life stress (ELS) is associated with adverse mental health outcomes including anxiety, depression and addiction-like behaviours. While ELS is known to affect the developing brain, leading to increased stress responsiveness and increased glucocorticoid levels, the molecular mechanisms underlying the detrimental effects of ELS remain incompletely characterised. Rodent models have been instrumental in beginning to uncover the molecular and cellular underpinnings of ELS. Limited nesting (LN), an ELS behavioural paradigm with significant improvements over maternal separation, mimics human maternal neglect. We have previously shown that LN leads to an increase in one of the behavioural measures of anxiety like-behaviours in rats (percent of entries in the EPM open arm). Here we assessed gene expression changes induced by ELS in rat prefrontal cortex by RNA-sequencing. We show that LN leads primarily to transcriptional repression and identify a molecular signature of LN in rat PFC that is observed across ELS protocols and replicable across rodent species (mouse and rat).
Collapse
Affiliation(s)
- Nicole Oldham Green
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jayanthi Maniam
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jessica Riese
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Irina Voineagu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
Simmons JM, Winsky L, Zehr JL, Gordon JA. Priorities in stress research: a view from the U.S. National Institute of Mental Health. Stress 2021; 24:123-129. [PMID: 32608314 DOI: 10.1080/10253890.2020.1781084] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mission of the National Institute of Mental Health is to transform the understanding and treatment of mental illnesses through basic and clinical research, paving the way for prevention, recovery, and cure. In consultation with a broad range of experts, the NIMH has identified a set of priorities for stress biology research aimed squarely at creating the basic and clinical knowledge bases for reducing and alleviating mental health burden across the lifespan. Here, we discuss these priority areas in stress biology research, which include: understanding the heterogeneity of stressors and outcomes; refining and expanding the experimental systems used to study stress and its effects; embracing and exploiting the complexity of the stress response; and prioritizing translational studies that seek to test mechanistic hypotheses in human beings. We emphasize the challenge of establishing mechanistic links across levels of analysis to explain how and when specific and diverse stressors lead to enduring changes in neural systems and produce lasting functional deficits in mental health relevant behaviors. An improved understanding of mechanisms underlying stress responses and the functional consequences of stress can and will speed translation from basic research to predictive markers of risk and to improved, personalized interventions for mental illness.
Collapse
Affiliation(s)
| | - Lois Winsky
- National Institute of Mental Health, Bethesda, MD, USA
| | - Julia L Zehr
- National Institute of Mental Health, Bethesda, MD, USA
| | | |
Collapse
|
28
|
Bis-Humbert C, García-Fuster MJ. Adolescent cocaine induced persistent negative affect in female rats exposed to early-life stress. Psychopharmacology (Berl) 2021; 238:3399-3410. [PMID: 34430991 PMCID: PMC8629899 DOI: 10.1007/s00213-021-05955-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023]
Abstract
RATIONALE The combination of several risk factors (sex, a prior underlying psychiatric condition, or early drug initiation) could induce the emergence of negative affect during cocaine abstinence and increase the risk of developing addiction. However, most prior preclinical studies have been centered in male rodents, traditionally excluding females from these analyses. OBJECTIVES To ascertain the behavioral and neurochemical consequences of adolescent cocaine exposure when the combination of several risk factors is present (female, early-life stress). METHODS Whole litters of Sprague-Dawley rats were exposed to maternal deprivation for 24 h on postnatal day (PND) 9. Cocaine was administered in adolescence (15 mg/kg/day, i.p., PND 33-39). Negative affect was assessed by several behavioral tests (forced swim, open field, novelty-suppressed feeding, sucrose preference). Hippocampal cell fate markers were evaluated by western blot (FADD, Bax, cytochrome c) or immunohistochemistry (Ki-67; cell proliferation). RESULTS Maternal deprivation is a suitable model of psychiatric vulnerability in which to study the impact of adolescent cocaine in female rats. While adolescent cocaine did not alter affective-like behavior during adolescence, a pro-depressive-like state emerged during adulthood, exclusively in rats re-exposed to cocaine during abstinence. FADD regulation by cocaine in early-life stressed female rats might contribute to certain hippocampal neuroadaptations with some significance to the observed induced negative affect. CONCLUSIONS Adolescent cocaine induced persistent negative affect in female rats exposed to early-life stress, highlighting the risk of early drug initiation during adolescence for the emergence of negative reinforcement during abstinence likely driving cocaine addiction vulnerability, also in female rats.
Collapse
Affiliation(s)
- Cristian Bis-Humbert
- grid.9563.90000 0001 1940 4767IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, 07122 Palma, Spain ,grid.507085.fHealth Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M. Julia García-Fuster
- grid.9563.90000 0001 1940 4767IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, 07122 Palma, Spain ,grid.507085.fHealth Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
29
|
A novel mouse model for vulnerability to alcohol dependence induced by early-life adversity. Neurobiol Stress 2020; 13:100269. [PMID: 33344722 PMCID: PMC7739069 DOI: 10.1016/j.ynstr.2020.100269] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/16/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022] Open
Abstract
Childhood adversity increases vulnerability to alcohol use disorders and preclinical models are needed to investigate the underlying neurobiological mechanisms. The present study modeled early-life adversity by rearing male and female C57BL/6J mouse pups in a limited bedding and nesting (LBN) environment, which induces erratic maternal care. As adults, mice were given limited access to two-bottle choice (2BC) alcohol drinking, combined or not with chronic intermittent ethanol (CIE) vapor inhalation to induce alcohol dependence. We tested the hypothesis that LBN rearing might exacerbate or facilitate the emergence of the motivational and affective effects of CIE. Consistent with our hypothesis, although LBN-reared males consumed the same baseline levels of alcohol as controls, they escalated their ethanol intake at an earlier stage of CIE exposure, i.e., after 4 rounds vs. 5 rounds for controls. In contrast, females were insensitive to both LBN rearing and CIE exposure. Males were further subjected to a behavioral test battery. Withdrawal from CIE-2BC increased digging activity and lowered mechanical nociceptive thresholds regardless of early-life conditions. On the other hand, LBN-reared CIE-2BC males showed reduced open arm exploration in the elevated plus maze and increased immobility in the tail suspension test compared to alcohol-naïve counterparts, while no group differences were detected among control-reared males. Finally, LBN rearing and alcohol exposure did not affect grooming in response to a sucrose spray (splash test), novel object recognition, or corticosterone levels. In summary, the LBN experience accelerates the transition from moderate to excessive alcohol drinking and produces additional indices of affective dysfunction during alcohol withdrawal in C57BL/6J male mice. Early-life adversity was generated by rearing C57BL/6J mouse pups in a limited bedding and nesting (LBN) environment. Alcohol dependence was induced in adulthood via chronic intermittent ethanol (CIE) inhalation. The LBN experience accelerated alcohol intake escalation in males. LBN exacerbated affective disturbances upon CIE withdrawal in males. Alcohol intake in females was insensitive to both LBN and CIE.
Collapse
|
30
|
Jardim NS, Müller SG, Pase FM, Nogueira CW. Nuclear Factor [Erythroid-derived 2]-like 2 and Mitochondrial Transcription Factor A Contribute to Moderate-intensity Swimming Effectiveness against Memory Impairment in Young Mice Induced by Concomitant Exposure to a High-calorie Diet during the Early Life Period. Neuroscience 2020; 452:311-325. [PMID: 33246070 DOI: 10.1016/j.neuroscience.2020.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/13/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Abstract
Increased energy food consumption during early-life has been associated with memory impairment. Swimming training has been reported to improve memory processes in rodent models. This study aimed to evaluate whether moderate-intensity swimming training counteracts learning and memory impairment in young mice fed a high-calorie diet during the early-life period. The contribution of hippocampal oxidative stress, as well as nuclear factor [erythroid-derived 2]-like 2/Kelch-like ECH-associated protein (NRF2/Keap-1/HO-1) and peroxisome proliferator-activated receptor gamma co-activator 1-alpha/mitochondrial transcription factor A (PCG-1α/mtTFA) signaling, in memory effects was also investigated. Three-week-old male Swiss mice received a high-calorie diet (20% fat; 20% carbohydrate enriched) or a standard diet from 21 to 49 postnatal days. Mice performed a moderate-intensity swimming protocol (5 days/week) and behavioral tests predictive of memory function. Mice fed a high-calorie diet and subjected to the swimming protocol performed better on short- and long-term spatial and object recognition memory tests than those fed a high-calorie diet. The swimming protocol modulated the hippocampal NRF2/Keap-1/HO-1 and mtTFA pathways in mice fed a high-calorie diet. Swimming training positively affected location and long-term memory, fat mass content, as well as NRF2/Keap-1/HO-1 and mtTFA proteins of control-diet-fed mice. In conclusion, a moderate-intensity swimming training evoked an adaptive response in mice fed a high-calorie diet by restoring different types of memory-impaired and hippocampal oxidative stress as well as upregulated the NRF2/Keap-1/HO-1 and mtTFA pathways.
Collapse
Affiliation(s)
- Natália Silva Jardim
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900 RS, Brazil
| | - Sabrina Grendene Müller
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900 RS, Brazil
| | - Flávia Matos Pase
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900 RS, Brazil
| | - Cristina Wayne Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900 RS, Brazil.
| |
Collapse
|
31
|
Hu P, Maita I, Phan ML, Gu E, Kwok C, Dieterich A, Gergues MM, Yohn CN, Wang Y, Zhou JN, Qi XR, Swaab DF, Pang ZP, Lucassen PJ, Roepke TA, Samuels BA. Early-life stress alters affective behaviors in adult mice through persistent activation of CRH-BDNF signaling in the oval bed nucleus of the stria terminalis. Transl Psychiatry 2020; 10:396. [PMID: 33177511 PMCID: PMC7658214 DOI: 10.1038/s41398-020-01070-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/15/2020] [Accepted: 10/07/2020] [Indexed: 01/06/2023] Open
Abstract
Early-life stress (ELS) leads to stress-related psychopathology in adulthood. Although dysfunction of corticotropin-releasing hormone (CRH) signaling in the bed nucleus of the stria terminalis (BNST) mediates chronic stress-induced maladaptive affective behaviors that are historically associated with mood disorders such as anxiety and depression, it remains unknown whether ELS affects CRH function in the adult BNST. Here we applied a well-established ELS paradigm (24 h maternal separation (MS) at postnatal day 3) and assessed the effects on CRH signaling and electrophysiology in the oval nucleus of BNST (ovBNST) of adult male mouse offspring. ELS increased maladaptive affective behaviors, and amplified mEPSCs and decreased M-currents (a voltage-gated K+ current critical for stabilizing membrane potential) in ovBNST CRH neurons, suggesting enhanced cellular excitability. Furthermore, ELS increased the numbers of CRH+ and PACAP+ (the pituitary adenylate cyclase-activating polypeptide, an upstream CRH regulator) cells and decreased STEP+ (striatal-enriched protein tyrosine phosphatase, a CRH inhibitor) cells in BNST. Interestingly, ELS also increased BNST brain-derived neurotrophic factor (BDNF) expression, indicating enhanced neuronal plasticity. These electrophysiological and behavioral effects of ELS were reversed by chronic application of the CRHR1-selective antagonist R121919 into ovBNST, but not when BDNF was co-administered. In addition, the neurophysiological effects of BDNF on M-currents and mEPSCs in BNST CRH neurons mimic effects and were abolished by PKC antagonism. Together, our findings indicate that ELS results in a long-lasting activation of CRH signaling in the mouse ovBNST. These data highlight a regulatory role of CRHR1 in the BNST and for BDNF signaling in mediating ELS-induced long-term behavioral changes.
Collapse
Affiliation(s)
- Pu Hu
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Isabella Maita
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Mimi L. Phan
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Edward Gu
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Christopher Kwok
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Andrew Dieterich
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Mark M. Gergues
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA ,grid.266102.10000 0001 2297 6811Present Address: Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Christine N. Yohn
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Yu Wang
- grid.59053.3a0000000121679639CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Hefei, 230027 China
| | - Jiang-Ning Zhou
- grid.59053.3a0000000121679639CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Hefei, 230027 China
| | - Xin-Rui Qi
- grid.412538.90000 0004 0527 0050Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Dick F. Swaab
- grid.418101.d0000 0001 2153 6865Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef, Amsterdam 1105 BA The Netherlands
| | - Zhiping P. Pang
- grid.430387.b0000 0004 1936 8796Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901 USA
| | - Paul J. Lucassen
- grid.7177.60000000084992262Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Troy A. Roepke
- grid.430387.b0000 0004 1936 8796Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA
| | - Benjamin A. Samuels
- grid.430387.b0000 0004 1936 8796Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| |
Collapse
|
32
|
Ding J, Chen X, da Silva MS, Lingeman J, Han F, Meijer OC. Effects of RU486 treatment after single prolonged stress depend on the post-stress interval. Mol Cell Neurosci 2020; 108:103541. [PMID: 32858150 DOI: 10.1016/j.mcn.2020.103541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022] Open
Abstract
The Single Prolonged Stress protocol is considered a model for PTSD, as it induces long lasting changes in rat behaviour and endocrine regulation. Previous work demonstrated that some of these changes can be prevented by treatment with the glucocorticoid receptor antagonist RU486, administered a week after the stressor. The current study evaluated the effects of an earlier intervention with RU486, as evaluated 1 week after SPS-exposure. Most RU486 effects occurred independent of prior stress, except for the reversal of a stress-induced increase in locomotor behaviour. The accompanying changes in gene expression depended on gene, brain region, and time. DNA methylation of the robustly down-regulated Fkbp5 gene was dissociated of changes in mRNA expression. The findings reinforce the long term effects of GR antagonist treatment, but also emphasize the need to evaluate changes over time to allow the identification of robust correlates between gene expression and behavioural/endocrine outcome of stressful experiences.
Collapse
Affiliation(s)
- Jinlan Ding
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands; PTSD Lab, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, PR China
| | - Xinzhao Chen
- PTSD Lab, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, PR China
| | - Marcia Santos da Silva
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Jolanthe Lingeman
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Fang Han
- PTSD Lab, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, PR China.
| | - Onno C Meijer
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
33
|
Cheng F, Cui S, Zhang C, Zhang L, Wang L, Yuan Q, Huang C, Zhang K, Zhou X. Association Between Cognitive Function and Early Life Experiences in Patients with Alcohol Use Disorder. Front Psychiatry 2020; 11:792. [PMID: 32903659 PMCID: PMC7438706 DOI: 10.3389/fpsyt.2020.00792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/23/2020] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Early life experiences could be potential risk factors for the development of alcohol use disorder. In similar circumstances, it might also influence cognitive impairment in later life. However, the relationship between early life experience and cognitive function in people with alcohol use disorders is unclear. The current study examined the effects of early social environments and experiences on cognitive function in patients with alcohol use disorder. METHODS A total of 37 adult male patients with alcohol use disorder and 30 healthy control (HC) subjects were enrolled. The MATRICS Cognitive Consensus Battery (MCCB) was used to evaluate cognitive function. The Childhood Trauma Questionnaire (CTQ) and the Revised Adult Attachment Scale (RAAS) were used to evaluate early life experiences of the participants. The RAAS was used to evaluate the attachment patterns of participants. RESULTS Compared with the HC group, the alcohol use disorder group reported higher levels of childhood abuse and were more likely to form an insecure attachment style. Patients with alcohol use disorder who experienced trauma performed worse in terms of discrete cognitive parameters such as social cognition, reasoning and problem solving compared to patients without trauma. Importantly, emotional neglect and social comfort were significantly associated with individual social cognitive skills. CONCLUSIONS Our results suggest that the cognitive function of patients with alcohol use disorder, especially social cognitive function, is affected by early life experiences.
Collapse
Affiliation(s)
- Fangshuo Cheng
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Shu Cui
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Chao Zhang
- Department of Pediatrics, Fuyang People's Hospital, Fuyang, China
| | - Ling Zhang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Lei Wang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Qiuyu Yuan
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Cui Huang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Kai Zhang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Xiaoqin Zhou
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, Hefei, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| |
Collapse
|
34
|
Malcon LMC, Wearick-Silva LE, Zaparte A, Orso R, Luft C, Tractenberg SG, Donadio MVF, de Oliveira JR, Grassi-Oliveira R. Maternal separation induces long-term oxidative stress alterations and increases anxiety-like behavior of male Balb/cJ mice. Exp Brain Res 2020; 238:2097-2107. [PMID: 32656651 DOI: 10.1007/s00221-020-05859-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/20/2020] [Indexed: 01/23/2023]
Abstract
Early life stress (ELS) exposure is a well-known risk factor for the development of psychiatric conditions, including anxiety disorder. Preclinical studies show that maternal separation (MS), a classical model of ELS, causes hypothalamic-pituitary-adrenal (HPA) axis alterations, a key contributor to the stress response modulation. Given that HPA axis activation has been shown to induce oxidative stress, it is possible to hypothesize that oxidative stress mediates the relationship between chronic ELS exposure and the development of several disorders. Here, we investigate the effects of MS in the oxidative status [plasma and brain reduced glutathione, catalase and thiobarbituric acid reactive substances (TBARS)], metabolism (glucose, triglycerides and cholesterol) and anxiety-like behaviors in adult Balb/cJ mice. In short, we found that MS increased anxiety-like behaviors in the open field, light/dark test but not in the elevated-plus maze. Animals also presented increased circulating cholesterol, increased TBARS in the plasma and decreased catalase in the hippocampus. Our findings suggest that MS induces long-term alterations in oxidative stress and increased anxiety-like behaviors.
Collapse
Affiliation(s)
- Luiza Martins Costa Malcon
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Luis Eduardo Wearick-Silva
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Aline Zaparte
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Rodrigo Orso
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Carolina Luft
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Saulo Gantes Tractenberg
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Márcio Vinicius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil. .,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
| |
Collapse
|
35
|
Wang D, Levine JLS, Avila-Quintero V, Bloch M, Kaffman A. Systematic review and meta-analysis: effects of maternal separation on anxiety-like behavior in rodents. Transl Psychiatry 2020; 10:174. [PMID: 32483128 PMCID: PMC7264128 DOI: 10.1038/s41398-020-0856-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/06/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
The mechanisms by which childhood maltreatment increases anxiety is unclear, but a propensity for increased defensive behavior in rodent models of early life stress (ELS) suggests that work in rodents may clarify important mechanistic details about this association. A key challenge in studying the effects of ELS on defensive behavior in rodents is the plethora of inconsistent results. This is particularly prominent with the maternal separation (MS) literature, one of the most commonly used ELS models in rodents. To address this issue we conducted a systematic review and meta-analysis, examining the effects of MS on exploratory-defensive behavior in mice and rats using the open field test (OFT) and the elevated plus maze (EPM). This search yielded a total of 49 studies, 24 assessing the effect of MS on behavior in the EPM, 11 tested behavior in the OFT, and 14 studies provided data on both tasks. MS was associated with increased defensive behavior in rats (EPM: Hedge's g = -0.48, p = 0.02; OFT: Hedge's g = -0.33, p = 0.05), effect sizes that are consistent with the anxiogenic effect of early adversity reported in humans. In contrast, MS did not alter exploratory behavior in mice (EPM: Hedge's g = -0.04, p = 0.75; OFT: Hedge's g = -0.03, p = 0.8). There was a considerable amount of heterogeneity between studies likely related to the lack of standardization of the MS protocol. Together, these findings suggest important differences in the ability of MS to alter circuits that regulate defensive behaviors in mice and rats.
Collapse
Affiliation(s)
- Daniel Wang
- grid.47100.320000000419368710Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
| | - Jessica L. S. Levine
- grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Victor Avila-Quintero
- grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Michael Bloch
- grid.47100.320000000419368710Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA ,grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
| |
Collapse
|
36
|
Berardino BG, Ballarini F, Chertoff M, Igaz LM, Cánepa ET. Nutritional stress timing differentially programs cognitive abilities in young adult male mice. Nutr Neurosci 2020; 25:286-298. [PMID: 32308155 DOI: 10.1080/1028415x.2020.1751507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objectives: The impact of chronic exposure to environmental adversities on brain regions involved in cognition and mental health depends on whether it occurs during the perinatal period, childhood, adolescence or adulthood. The effects of these adversities on the brain and behavior arise as a function of the timing of the exposure and their co-occurrence with the development of specific regions. Here we aimed to explore the behavioral phenotypes derived from two nutritional stress paradigms which differed in the timing of exposure: a low-protein perinatal diet during gestation and lactation and a low-protein diet during adolescence.Methods: Locomotor and exploratory activity, recognition memory and aversive memory were measured in CF-1 8-week-old male mice subjected to perinatal malnutrition (LP-P) or adolescent malnutrition (LP-A), and their respective controls with normal protein diet (NP-P and NP-A).Results: By using the open field test, we found that LP-P and LP-A mice showed reduced exploratory activity compared to controls, but no alterations in their locomotor activity. Recognition memory was impaired only in LP-P mice. Interestingly, aversive memory was not altered in LP-P mice but was enhanced in LP-A mice. Considering the stress-inoculation theory, we hypothesized that protein malnutrition during adolescence represents a challenging but still moderate stressful environment, which promotes active coping in face of later adversity.Conclusion: Our results indicate that while perinatal malnutrition impairs recognition memory, adolescent malnutrition enhances aversive memory, showing dissimilar adaptive responses.
Collapse
Affiliation(s)
- Bruno G Berardino
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de Ciencias Exactas y Naturales, CONICET, Ciudad de Buenos Aires, Argentina
| | - Fabricio Ballarini
- Facultad de Medicina, Universidad de Buenos Aires - IBCN "Eduardo De Robertis" (CONICET), Ciudad de Buenos Aires, Argentina
| | - Mariela Chertoff
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de Ciencias Exactas y Naturales, CONICET, Ciudad de Buenos Aires, Argentina
| | - Lionel M Igaz
- IFIBIO Bernardo Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET, Buenos Aires, Argentina
| | - Eduardo T Cánepa
- Laboratorio de Neuroepigenética, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, and Instituto de Química Biológica de Ciencias Exactas y Naturales, CONICET, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
37
|
Synthesizing Views to Understand Sex Differences in Response to Early Life Adversity. Trends Neurosci 2020; 43:300-310. [PMID: 32353334 DOI: 10.1016/j.tins.2020.02.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/11/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Sex as a biological variable (SABV) is critical for understanding the broad range of physiological, neurobiological, and behavioral consequences of early life adversity(ELA). The study of the interaction of SABV and ELA ties into several current debates, including the importance of taking into account SABV in research, differing strategies employed by males and females in response to adversity, and the possible evolutionary and developmental mechanisms of altered development in response to adversity. This review highlights the importance of studying both sexes, of understanding sex differences (and similarities) in response to ELA, and provides a context for the debate surrounding whether the response to ELA may be an adaptive process.
Collapse
|
38
|
Lucassen PJ, Fitzsimons CP, Salta E, Maletic-Savatic M. Adult neurogenesis, human after all (again): Classic, optimized, and future approaches. Behav Brain Res 2020; 381:112458. [DOI: 10.1016/j.bbr.2019.112458] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/29/2019] [Accepted: 12/28/2019] [Indexed: 02/08/2023]
|
39
|
Pittaras E, Rabat A, Granon S. The Mouse Gambling Task: Assessing Individual Decision-making Strategies in Mice. Bio Protoc 2020; 10:e3479. [PMID: 33654712 DOI: 10.21769/bioprotoc.3479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/22/2019] [Accepted: 11/26/2019] [Indexed: 11/02/2022] Open
Abstract
Decision-making is a complex cognitive process which consists of choosing one option among several alternatives. In humans, this process is featured in the Iowa gambling task (IGT), a decision-making task that mimics real life situations by reproducing uncertain conditions based on probabilistic rewards or penalties (see Background). Several authors wanted to adapt the IGT in rodents with subtle differences in protocols that match various aspects of the human task. Here we propose, for the first time in mice, a protocol that contains the most important characteristics of the IGT: 4 different options, choices based on 4 ambiguous outcomes with immediate and long term rewards, a total of 100 trials, no learning of the contingency before the task, and presence of both a certain reward and a probable penalty. During this task, mice have to choose between options more or less advantageous in the short and long term by developing a decision-making strategy that differs between individuals. Therefore, the strength of this protocol is that it is one of the first to enable the study of decision-making in a complex situation, and demonstrates inter-individual differences regarding decision-making strategies in mice.
Collapse
Affiliation(s)
- Elsa Pittaras
- Biology Department, Stanford University, Stanford, CA 94305-5020, USA
| | - Arnaud Rabat
- Unité Fatigue et Vigilance, Département Environnements Opérationnels, Institut de Recherche Biomédicale des armées, 1 place du Général Valérie ANDRE, BP 73, 91223 Bretigny-sur-Orge cedex, France.,Equipe d'accueil 7330 «VIFASOM», Hôtel Dieu AP-HP, Université Paris 5 Descartes, 1 place du Parvis Notre Dame, 75181 Paris cedex 04, France
| | - Sylvie Granon
- Neurobiology of decision making, Paris-Saclay Institute of Neuroscience (Neuro-PSI), CNRS UMR 9197, 91400 Orsay, France
| |
Collapse
|
40
|
Agarwal P, Palin N, Walker SL, Glasper ER. Sex-dependent effects of paternal deprivation and chronic variable stress on novel object recognition in adult California mice (Peromyscus californicus). Horm Behav 2020; 117:104610. [PMID: 31669457 DOI: 10.1016/j.yhbeh.2019.104610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
Early-life stress exposure can confer vulnerability for development of psychiatric illnesses and impaired cognition in adulthood. It is well-known that early-life stress can dysregulate the hypothalamic-pituitary-adrenal (HPA) axis in a sex-dependent manner. Specifically, uniparental rodent models of prolonged disrupted mother-offspring relationships (e.g., maternal separation) have demonstrated greater alterations in stress responsivity in adult males, compared to females. Also, chronic early-life stressors (e.g., limited bedding model) impair cognitive function in males more than females. However, the sex-dependent effects of early-life stress and later-life chronic HPA axis activation on cognition have not been well-characterized. Here, we utilized the biparental California mouse (Peromyscus californicus) to model the early-life adversity of paternal deprivation (PD). Fathers either remained in the nest (biparental care) or were permanently removed (PD) on postnatal day (PND) 1. Adult offspring were exposed to daily handling (control) or chronic variable stress (CVS; three stressors for seven days). Twenty-four hours after the final stressor, the novel object recognition (NOR) task commenced, followed by serum collection for corticosterone (CORT) analysis. Independent of sex or rearing, CVS increased CORT. Exploration during acquisition for the NOR task was increased as a result of CVS and PD. During NOR testing, non-stressed females exhibited greater difference scores (i.e., increased recognition memory), compared to non-stressed males. However, the addition of CVS diminished difference scores in females - an effect not observed in CVS-exposed males. Overall, these data suggest that neonatal paternal experience, sex, and chronic stress contribute to exploratory behavior, cognition, and stress hormone concentrations in a biparental species.
Collapse
Affiliation(s)
- P Agarwal
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - N Palin
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - S L Walker
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742, USA
| | - E R Glasper
- Department of Psychology, University of Maryland, College Park, MD 20742, USA; Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
41
|
Zhang X, Li H, Sun H, Jiang Y, Wang A, Kong Y, Sun X, Zhu G, Li Q, Du Z, Sun H, Sun L. Effects of BDNF Signaling on Anxiety-Related Behavior and Spatial Memory of Adolescent Rats in Different Length of Maternal Separation. Front Psychiatry 2020; 11:709. [PMID: 32793001 PMCID: PMC7391957 DOI: 10.3389/fpsyt.2020.00709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
As an adverse form of early-life stress (ELS), maternal separation (MS) can interfere with the development of cognition and behaviors of adolescent rodents. Brain-derived neurotrophic factor (BDNF) is involved in the regulation of brain development and function, but the molecular mechanisms by which BDNF regulates brain function and behavior in MS with different stressor strengths remain unclear. This descriptive study characterized the levels of BDNF in the prefrontal cortex (PFC) and plasma corticosterone (CORT) from the offspring of rats exposed to early handling (EH, 15-min separation per day) and prolonged MS (PMS, 180-min separation per day), during postnatal days (PND) 1‑21. The behavioral and biochemical analyses were performed during adolescence (PND 42‑56). PMS resulted in reduced weight and decreased locomotor activity in the open field test and Y-maze task compared to control (CON) group, with EH showing an intermediate phenotype. BDNF protein levels in the PFC were lower in PMS compared to EH and further reduced in CON male rats. Plasma CORT levels were higher in PMS compared to CON with EH again showing intermediate levels. Neither PMS or EH affected spatial learning in the Y-maze task. These findings indicate that longer periods of maternal separation are necessary to increase anxiety-like behavior, elevate CORT levels, and further suppress BDNF levels in the PFC, providing a possible mechanism to explain why more severe forms of ELS lead to more significant psychiatric and medical consequences later in life.
Collapse
Affiliation(s)
- Xianqiang Zhang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Haonan Li
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Haoran Sun
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yinghong Jiang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Aihong Wang
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yujia Kong
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Xiue Sun
- Department of Nursing, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guohui Zhu
- Depression Treatment Center, Weifang Mental Health Center, Weifang, China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, Hong Kong
| | - Zhongde Du
- Department of Neurology, Sunshine Union Hospital, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Lin Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| |
Collapse
|
42
|
Qu N, Zhang SF, Xia B, Xie JZ, Wang XM, Liu J, Xiong J, Ren N, Wang Y, Tian Q, Li Y. Sex difference in IL-6 modulation of cognition among Chinese individuals with major depressive disorder. J Clin Neurosci 2019; 70:14-19. [DOI: 10.1016/j.jocn.2019.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/05/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
|
43
|
Abstract
In the past several years, there has been an explosion of interest in animal models of risk-based decision-making, a fundamental process associated with gambling disorder. While early work focused on establishing various tasks for assaying decision-making, current studies are determining the (subtle and not-so-subtle) influence of cues in driving risky decisions to better understand problem gambling. In addition, these decision-making paradigms are now being used to investigate comorbid conditions such as substance dependence or brain injury and replicating observations from human patients. These animal models have now developed to a point where therapeutic interventions may be assessed for not just gambling disorder, but also a number of other conditions which engender risky decision-making.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Injury and Recovery Laboratory.,Department of Psychology, West Virginia University, Morgantown, WV, USA.,Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
44
|
Kokras N, Hodes GE, Bangasser DA, Dalla C. Sex differences in the hypothalamic-pituitary-adrenal axis: An obstacle to antidepressant drug development? Br J Pharmacol 2019; 176:4090-4106. [PMID: 31093959 PMCID: PMC6877794 DOI: 10.1111/bph.14710] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 12/30/2022] Open
Abstract
Hypothalamic-pituitary-adrenal (HPA) axis dysfunction has long been implicated in the pathophysiology of depression, and HPA axis-based compounds have served as potential new therapeutic targets, but with no success. This review details sex differences from animal and human studies in the function of HPA axis elements (glucocorticoids, corticotropin releasing factor, and vasopressin) and related compounds tested as candidate antidepressants. We propose that sex differences contribute to the failure of novel HPA axis-based drugs in clinical trials. Compounds studied preclinically in males were tested in clinical trials that recruited more, if not exclusively, women, and did not control, but rather adjusted, for potential sex differences. Indeed, clinical trials of antidepressants are usually not stratified by sex or other important factors, although preclinical and epidemiological data support such stratification. In conclusion, we suggest that clinical testing of HPA axis-related compounds creates an opportunity for targeted, personalized antidepressant treatments based on sex. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Nikolaos Kokras
- Department of PharmacologyNational and Kapodistrian University of AthensAthensGreece
- First Department of Psychiatry, Eginition HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Georgia E. Hodes
- School of NeuroscienceVirginia Polytechnic Institute and State UniversityBlacksburgVirginia
| | | | - Christina Dalla
- Department of PharmacologyNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
45
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
46
|
Nisar S, Farooq RK, Nazir S, Alamoudi W, Alhibshi A. Exposure to early life adversity alters the future behavioral response to a stressful challenge in BALB/C mice. Physiol Behav 2019; 210:112622. [PMID: 31325513 DOI: 10.1016/j.physbeh.2019.112622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/19/2019] [Accepted: 07/16/2019] [Indexed: 12/01/2022]
Abstract
Depression is considered as a maladaptive response to stress in adult life. Exposure to stress in early childhood is recognized as a risk factor for being unable to adapt to environmental changes in adult life. Early life stress (ELS) has been modelled in animals to help understand the behavioral outcome of the adversity. Periodic maternal separation (MS) in rodents for the first two weeks of life is one such model. We used MS as a form of ELS in Balb/c mice to study its effect on a stressful challenge encountered in adult life. According to our results, exposure to MS predisposed mice to an altered behavioral response. However, this response was not worsened by exposure to restraint stress (RS) experienced in early adult life. This controversy may be attributed to methodological and biological variations among animals as well as humans.
Collapse
Affiliation(s)
- Safia Nisar
- Pakistan Biological Safety Association PBSA, Pakistan
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O Box 1982, Dammam 31441, Saudi Arabia.
| | - Sadia Nazir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Widyan Alamoudi
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O Box 1982, Dammam 31441, Saudi Arabia
| | - Amani Alhibshi
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
47
|
White JD, Kaffman A. The Moderating Effects of Sex on Consequences of Childhood Maltreatment: From Clinical Studies to Animal Models. Front Neurosci 2019; 13:1082. [PMID: 31680821 PMCID: PMC6797834 DOI: 10.3389/fnins.2019.01082] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Stress has pronounced effects on the brain, and thus behavioral outputs. This is particularly true when the stress occurs during vulnerable points in development. A review of the clinical literature regarding the moderating effects of sex on psychopathology in individuals exposed to childhood maltreatment (CM) is complicated by a host of variables that are difficult to quantify and control in clinical settings. As a result, the precise role of sex in moderating the consequences of CM remains elusive. In this review, we explore the rationale for studying this important question and their implications for treatment. We examine this issue using the threat/deprivation conceptual framework and highlight a growing body of work demonstrating important sex differences in human studies and in animal models of early life stress (ELS). The challenges and obstacles for effectively studying this question are reviewed and are followed by recommendations on how to move forward at the clinical and preclinical settings. We hope that this review will help inspire additional studies on this important topic.
Collapse
Affiliation(s)
- Jordon D White
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Arie Kaffman
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
48
|
Klinger K, Gomes FV, Rincón-Cortés M, Grace AA. Female rats are resistant to the long-lasting neurobehavioral changes induced by adolescent stress exposure. Eur Neuropsychopharmacol 2019; 29:1127-1137. [PMID: 31371105 PMCID: PMC6773464 DOI: 10.1016/j.euroneuro.2019.07.134] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/30/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023]
Abstract
Stress during adolescence is a risk factor for neuropsychiatric diseases, including schizophrenia. We recently observed that peripubertal male rats exposed to a combination of daily footshock plus restraint stress exhibited schizophrenia-like changes. However, numerous studies have shown sex differences in neuropsychiatric diseases as well as on the impact of coping with stress. Thus, we decided to evaluate, in adolescent female rats, the impact of different stressors (restraint stress [RS], footshock [FS], or the combination of FS and RS [FS+RS]) on social interaction (3-chamber social approach test/SAT), anxiety responses (elevated plus-maze/EPM), cognitive function (novel object recognition test/NOR), and dopamine (DA) system responsivity by evaluating locomotor response to amphetamine and in vivo extracellular single unit recordings of DA neurons in the ventral tegmental area (VTA) in adulthood. The impact of FS+RS during early adulthood was also investigated. Adolescent stress had no impact on social behavior, anxiety, cognition and locomotor response to amphetamine. In addition, adolescent stress did not induce long-lasting changes in VTA DA system activity. However, a decrease in the firing rate of VTA DA neurons was found 1-2 weeks post-adolescent stress. Similar to adolescent stress, adult stress did not induce long-lasting behavioral deficits and changes in VTA DA system activity, but FS+RS decreased VTA DA neuron population activity 1-2 weeks post-adult stress. Our results are consistent with previous studies showing that female rodents appear to be more resilient to developmental stress-induced persistent changes than males and may contribute to the delayed onset and lesser severity of schizophrenia in females.
Collapse
Affiliation(s)
- Katharina Klinger
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Institute of Genetic and Molecular Neurobiology, Otto-von-Guericke University, University of Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Felipe V Gomes
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, 3900 Bandeirantes Ave, Ribeirao Preto, SP, 14049-900, Brazil
| | - Millie Rincón-Cortés
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
49
|
Caruso A, Nicoletti F, Gaetano A, Scaccianoce S. Risk Factors for Alzheimer's Disease: Focus on Stress. Front Pharmacol 2019; 10:976. [PMID: 31551781 PMCID: PMC6746823 DOI: 10.3389/fphar.2019.00976] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
In vulnerable individuals, chronic and persistent stress is an established risk factor for disorders that are comorbid with Alzheimer’s disease (AD), such as hypertension, obesity and metabolic syndrome, and psychiatric disorders. There are no disease-modifying drugs in the treatment of AD, and all phase-3 clinical trials with anti-amyloid drugs (e.g., β- or γ-secretase inhibitors and monoclonal antibodies) did not meet the primary endpoints. There are many reasons for the lack of efficacy of anti-amyloid drugs in AD, the most likely being a late start of treatment, considering that pathophysiological mechanisms underlying synaptic dysfunction and neuronal death begin several decades before the clinical onset of AD. The identification of risk factors is, therefore, an essential step for early treatment of AD with candidate disease-modifying drugs. Preclinical studies suggest that stress, and the resulting activation of the hypothalamic–pituitary–adrenal axis, can induce biochemical abnormalities reminiscent to those found in autoptic brain samples from individuals affected by AD (e.g., increases amyloid precursor protein and tau hyperphosphorylation). In this review, we will critically analyze the current knowledge supporting stress as a potential risk factor for AD.
Collapse
Affiliation(s)
- Alessandra Caruso
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy.,Neuropharmacology Research Unit, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Alessandra Gaetano
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
50
|
Keller SM, Nowak A, Roth TL. Female pups receive more maltreatment from stressed dams. Dev Psychobiol 2019; 61:824-831. [PMID: 30810229 PMCID: PMC6711830 DOI: 10.1002/dev.21834] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 01/10/2023]
Abstract
The effects of exposure to developmental stress often diverge for males and females. Using the scarcity-adversity model of low nesting resources outside the home cage, our lab has discovered sex differences in both behavioral and epigenetic consequences of repeated exposure to caregiver maltreatment. For the measures we have performed to date, we have found more consequences for females. The reasons underlying this sex disparity are unknown. In the current experiment, we aimed to discern the quality of maternal care received by male and female pups in our model. As we have previously found more behavioral and epigenetic consequences in females, we hypothesized that females receive more adverse care compared to their male littermates. Our hypothesis was supported; in our maltreatment condition, we found that female pups received more adverse care than males. This sex difference in adverse care was not present in our two control conditions (cross-foster and normal maternal care). These data lend support to the notion that one reason females in our model incur more behavioral and epigenetic consequences is a result of greater mistreatment by the dam.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Anna Nowak
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|