Elsevier

Hormones and Behavior

Volume 100, April 2018, Pages 69-80
Hormones and Behavior

Multimodal early-life stress induces biological changes associated to psychopathologies

https://doi.org/10.1016/j.yhbeh.2018.03.005Get rights and content

Highlights

  • Physical and psychological stressors early in life promote changes in corticosterone response.

  • Early life stress acutely elicits HPA axis activity and induces persistent behavioral alterations.

  • Anxiety and depressive-like phenotype induced by ELS is dissociated from hippocampal plasticity.

Abstract

Evidences suggest the contributive role of early-life stress (ELS) to affective and anxiety disorders. Chronic exposure to the same stressor may generate habituation, while the exposure to different and repeated stressors gradually promotes maladaptive plasticity. Therefore, to further understand the effects of heterotypic stressors during early life period, male Wistar rat pups (P1–P21) were exposed to Multimodal ELS paradigm. Results indicate pups did not habituate to multimodal ELS and neonates respond to both physical and psychogenic stressors. Adult rats that underwent ELS protocol showed significant lower sucrose intake, decreased latency to immobility in the forced swim test and increased latency to light compartment in the light-dark test when compared to control group. Although it has been shown that ELS-induced changes in hippocampus can be used as biomarkers, multimodal ELS did not significantly alter BDNF, Tyrosine Kinase B (TrkB) receptor expression or neurogenesis in the hippocampus. Taken together, these findings indicate that multimodal ELS protocol can be an interesting experimental model for understanding long-term psychiatric disorders associated with stress. Indeed, our data with neurogenesis, BDNF and TrkB, and conflicting data from the literature, suggest that additional studies on synaptic plasticity/intracellular cascades would help to detect the underlying mechanisms.

Introduction

Adversities in early life are associated with vulnerability to psychopathologies later in life, resulting in long-term impact in emotional function (Chrousos, 2009; Gold et al., 1988; Heim and Nemeroff, 2002; Juruena, 2013; Lippmann et al., 2007; Lupien et al., 2009).

Patients suffering with stress-related psychiatric disorders appear to be especially sensitive to the effects of early life stress (ELS), which is supported by epidemiological data (Baes et al., 2014; Juruena, 2013). For instance, traumatic childhood experiences such as abuse, neglect and parental losses increase the incidence of Major Depression (MD) (Anda et al., 2006) which could reach in adult life between 59% and 75% (Widom et al., 2007). Depression affects millions of people and causes significant impairments on patient's quality of life (Ustün et al., 2004). Epidemiological data ranks depression in the top fourth causes of global burden of disease (Mathers and Loncar, 2006; Murray and Lopez, 1997), thus representing a serious problem of public health worldwide. Projections estimate that in 2020 nearly one in ten people on the planet (676 million) will present a depressive episode (World Health Organization, 2017).

Anxiety disorders are often observed as comorbid with depression, indicating common neurobiological mechanisms between these mental illnesses (Craske et al., 2009; Craske and Stein, 2016). People that experience early emotional traumas are 1.9 to 3.6 fold more likely to develop anxiety disorders (Fernandes and Osório, 2015). Depression-anxiety comorbidity is strongly associated with impairment in health, as well as in emotional functions (Kroenke et al., 2007).

A better understanding of how stress in early life impacts brain and behavior in adulthood is, therefore, of fundamental importance to the study of the neurobiology of psychiatric disorders. Although plenty of evidences indicate that ELS exposure leads to behavioral changes in adulthood (Bale et al., 2011; Gee and Casey, 2015; Lai and Huang, 2011; Loi et al., 2014; Lupien et al., 2009; Walker et al., 2017), there are some reports proposing that there is a period during early life in which the response to stress is reduced or absent, also known as the stress hyporesponsive period (SHRP). Interestingly, ELS experiences can disrupt the Stress SHRP, turning the HPA-axis responsive to stressors (Daskalakis et al., 2015) and there is growing evidence that brain functioning during development is affected by ELS (Cowan et al., 2016). In light of these data the use of animal models can provide a better understanding of the pathological mechanisms that increases vulnerability to depression and other stress-related psychiatry disorders, since the relationship between environmental factors, as well as the underlying brain mechanisms involved in behavioral changes, may be better examined (Abelaira et al., 2013).

A number of animal models have been developed to investigate the underlying changes caused by ELS on the developing brain. Since the 1970's, studies that started with Levine and colleagues have demonstrated that early developmental manipulations related to maternal care, commonly, by maternal separation (MS) paradigm, result in intermittent stress and provoke a profound and lasting impact on emotionality and stress response (Levine, 1967; Levine, 2005; Nishi et al., 2014). Since then, most ELS long-lasting effects were obtained by employing protocols that require long periods of separation (with daily separation for 3 h, or maternal deprivation for 24 h) or a second hit (Holmes et al., 2005). But only a few studies have shown significant structural changes in limbic brain regions, in the HPA axis and in behavior in adulthood (Loi et al., 2014).

Therefore, rodent models findings have been supported upon protocols based on one type of psychological stressor, altering the amount of maternal care, which may not necessarily reflect a translational perspective. Even though MS models have provided a vast amount of data on the effects of reduced maternal input on pup development, this manipulation may differ from the human condition since the mother is typically present but infants and children grow up in chronic rather than intermittent stress (Molet et al., 2014). Moreover, the odds of developing depressive disorders with exposure to physical stressors were greatest in prospective studies (Norman et al., 2012). Chronic variable stress protocol, which applies different modalities of stressors, is commonly employed in adult life. Although this type of paradigm is extensively used, its impacts have not been studied early in life.

Development of brain structures related to stress system seems to be tuned specifically to modalities relevant to the early-life period. Also, the chronic exposure to the same stressor may generate habituation (Gadek-Michalska & Bugajski, 2003) while the exposure to different and repeated stressors gradually changes the electrical, proliferative and morphological characteristics of neurons (Joëls et al., 2007). Therefore, protocols employing chronic variable stress during early life could provide a valuable tool to investigate stress effects on neurodevelopment and behavior in adulthood. Although this type of paradigm is extensively used in adults (Willner, 2017), its impacts have not been studied early in life.

There is mounting evidence of multiple dysregulated mechanisms that can be considered interesting biomarkers but reduced levels of growth factors and altered endocrine/metabolic function stand out (Schmidt et al., 2011). Although neurotrophic factors are known to play an important role in the growth and survival of neurons (Mpofana et al., 2016), the role of brain derived neurotrophic factor (BDNF) or its main receptor, tyrosine kinase B (TrkB) in the pathophysiology of depression is in fact complex and not completely understood (Castrén and Rantamäki, 2010). Nevertheless, a clear direction (increase vs decrease) of the expression of either hippocampal BDNF or TrkB after ELS has not been detected (Daskalakis et al., 2015), neither it has been reported a distinction between dorsal and ventral hippocampal sub-regions. Additionally to BDNF-TrkB signaling, disturbed adult neurogenesis may also contribute to hippocampus malfunctioning and symptom development in depressed patients (Lee et al., 2013).

Therefore, it is conceivable that the controllability and predictability of stimuli are important developmental determinants for the adult capacity to cope with environmental demands (Koolhaas et al., 2011). Considering this, our study investigated the effects of a new variable protocol of early life stress on behavioral and molecular changes related to depression, later in adulthood, with the aim to provide a new framework for understanding how changes in neuroendocrine function and their relationship with depression and anxiety behavior can be better explored.

Section snippets

Animals

Female rats between 18 and 21 days of pregnancy were acquired from the Central Vivarium of the University of São Paulo, Ribeirão Preto Campus (USP-RP) and monitored twice a day until birth. Animals were housed in transparent acrylic boxes (45x32x17cm), at the Animal Facility of the Physiology Department of the Ribeirão Preto School of Medicine at the University of São Paulo, under controlled ventilation and temperature (25 ± 2 °C) with light/dark cycle of 12 h (lights on 7:00 A.M.) and with

Experiment 1 – different effects of acute stressors during early life on corticosterone plasmatic concentration

Different modalities of acute stressors (Fig. 1) on early life period promoted different responses on plasmatic corticosterone secretion (ANOVA one-way F(6,53) = 5.702; p < 0.0001; r2 = 0,03923). We found that from all the stressors tested, 10 min of Maternal Separation (p < 0.0003), 60 min of Restraint with Agitation (p < 0.0006), Injection (p < 0.0027) and Cold exposure (p < 0.0003) were able to significantly increase the plasmatic concentration of corticosterone in PND12, respectively

Short-term effects of different acute stressors

We observed increased plasmatic corticosterone levels when different types of acute stressors are presented for 10 min in PND12, except for restraint with agitation. Also, in our study increased corticosterone level does not remain significantly elevated 30 min after the end of 60 min of the maternal separation procedure.

Although there are reports of increased corticosterone after MS (different ages or different duration), our data are in line with other studies, such as McCormick et al., 1998,

Conclusions

Our findings demonstrate that during the Multimodal ELS paradigm exposure, the HPA axis is activated and there is no habituation to stressors. Also, data support that, during early life period, animals respond differently to stressor modalities over time. In fact, our current data indicate that this ELS paradigm is effective to induce, later in life, alterations in anhedonia, depressive and anxiety-like behavior and HPA axis functioning, in spite of the fact that we did not observe changes in

Acknowledgments

The research was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo – FAPESP (LDG 2017/11339-0, 2014/17959-1, EHLU 2015/18773-1, DER 2013/01737-7), Conselho Nacional de Pesquisa e Desenvolvimento – CNPq (NGC Investigator Fellowship) and Coordenadoria de Aperfeiçoamento de Pessoal de Nível Superior – PROEX-CAPES Project for the Graduate Program in Physiology of the FMRP-USP.

References (85)

  • A. Holmes et al.

    Early life genetic, epigenetic and environmental factors shaping emotionality in rodents

  • G.-J. Huang et al.

    The role of 5-HT1A receptors in the proliferation and survival of progenitor cells in the dentate gyrus of the adult hippocampus and their regulation by corticoids

    Neuroscience

    (2005)
  • M. Joëls et al.

    Chronic stress: implications for neuronal morphology, function and neurogenesis

    Front. Neuroendocrinol.

    (2007)
  • N. Kee et al.

    The utility of Ki-67 and BrdU as proliferative markers of adult neurogenesis

    J. Neurosci. Methods

    (2002)
  • J.M. Koolhaas et al.

    Stress revisited: a critical evaluation of the stress concept

    Neurosci. Biobehav. Rev.

    (2011)
  • M. Lai et al.

    Effects of early life stress on neuroendocrine and neurobehavior: mechanisms and implications

    Pediatr. Neonatol.

    (2011)
  • S. Levine

    Developmental determinants of sensitivity and resistance to stress

    Psychoneuroendocrinology

    (2005)
  • C. Lino-de-Oliveira et al.

    Structure of the rat behaviour in the forced swimming test

    Behav. Brain Res.

    (2005)
  • R. Llorente et al.

    Early maternal deprivation and neonatal single administration with a cannabinoid agonist induce long-term sex-dependent psychoimmunoendocrine effects in adolescent rats

    Psychoneuroendocrinology

    (2007)
  • C.M. McCormick et al.

    Corticosterone release in response to repeated, short episodes of neonatal isolation: evidence of sensitization

    Int. J. Dev. Neurosci.

    (1998)
  • M.L. Molendijk et al.

    Immobility in the forced swim test is adaptive and does not reflect depression

    Psychoneuroendocrinology

    (2015)
  • C.J. Murray et al.

    Global mortality, disability, and the contribution of risk factors: global burden of disease study

    Lancet

    (1997)
  • S. Ortega-Martinez

    Influences of prenatal and postnatal stress on adult hippocampal neurogenesis: the double neurogenic niche hypothesis

    Behav. Brain Res.

    (2015)
  • S.G. Tractenberg et al.

    An overview of maternal separation effects on behavioural outcomes in mice: evidence from a four-stage methodological systematic review

    Neurosci. Biobehav. Rev.

    (2016)
  • E.H.L. Umeoka et al.

    Functional characterization of the hypothalamic-pituitary-adrenal axis of the Wistar Audiogenic Rat (WAR) strain

    Brain Res.

    (2011)
  • A. Wigger et al.

    Periodic maternal deprivation induces gender-dependent alterations in behavioral and neuroendocrine responses to emotional stress in adult rats

    Physiol. Behav.

    (1999)
  • P. Willner

    Neurobiology of stress the chronic mild stress (CMS) model of depression: history, evaluation and usage

    Neurobiol. Stress

    (2017)
  • J. Zhang et al.

    Behavioral deficits, abnormal corticosterone, and reduced prefrontal metabolites of adolescent rats subject to early life stress

    Neurosci. Lett.

    (2013)
  • H.M. Abelaira et al.

    Animal models as tools to study the pathophysiology of depression

    Rev. Bras. Psiquiatr.

    (2013)
  • J. Altman et al.

    Migration and distribution of two populations of hippocampal granule cell precursors during the perinatal and postnatal periods

    J. Comp. Neurol.

    (1990)
  • R.F. Anda et al.

    The enduring effects of abuse and related adverse experiences in childhood: a convergence of evidence from neurobiology and epidemiology

    Eur. Arch. Psychiatry Clin. Neurosci.

    (2006)
  • C. von W. Baes et al.

    Early life stress in depressive patients: HPA axis response to GR and MR agonist

    Front. Psych.

    (2014)
  • T.L. Bale et al.

    Early life programming and neurodevelopmental disorders Tracy

    Biol. Psychiatry

    (2011)
  • J. Bugajski

    Repeated Handling, Restraint, or Chronic Crowding Impair the Hypothalamic-pituitary-adrenocortical Response to Acute Restraint Stress

    (2003)
  • F.L.M. Cardenas et al.

    X-Plo-Rat: A Free Software for Behavior Recording

    (2001)
  • E. Castrén et al.

    The role of BDNF and its receptors in depression and antidepressant drug action: reactivation of developmental plasticity

    Dev. Neurobiol.

    (2010)
  • O.W. Castro et al.

    Impact of Corticosterone Treatment on Spontaneous Seizure Frequency and Epileptiform Activity in Mice with Chronic Epilepsy

    (2012)
  • G.P. Chrousos

    Stress and disorders of the stress system

    Nat. Rev. Endocrinol.

    (2009)
  • C.S.M. Cowan et al.

    The lasting impact of early-life adversity on individuals and their descendants: potential mechanisms and hope for intervention

    Genes Brain Behav.

    (2016)
  • N.P. Daskalakis et al.

    Early life stress effects on glucocorticoid—BDNF interplay in the Hippocampus

    Front. Mol. Neurosci.

    (2015)
  • C.V. Dayas et al.

    Neuroendocrine responses to an emotional stressor: evidence for involvement of the medial but not the central amygdala

    Eur. J. Neurosci.

    (1999)
  • C.V. Dayas et al.

    Stressor categorization: acute physical and psychological stressors elicit distinctive recruitment patterns in the amygdala and in medullary noradrenergic cell groups

    Eur. J. Neurosci.

    (2001)
  • Cited by (0)

    View full text