Elsevier

Brain Research Bulletin

Volume 172, July 2021, Pages 164-179
Brain Research Bulletin

Chronic unpredictable stress negatively regulates hippocampal neurogenesis and promote anxious depression-like behavior via upregulating apoptosis and inflammatory signals in adult rats

https://doi.org/10.1016/j.brainresbull.2021.04.017Get rights and content

Highlights

  • Enhanced glial activation and neuroinflammation cause anxious depression in rats after CUS exposure.

  • CUS mediated reduced adult hippocampal neurogenesis results in anxious depression.

  • Increased apoptosis is observed in cortex and hippocampus of rat brain after CUS exposure.

  • Downregulated p-Akt/Akt and upregulated p-Erk/Erk ratio may cause anxious depression.

Abstract

Psychological and physical stress play a pivotal role in etiology of anxiety and depression. Chronic psychological and physical stress modify various physiological phenomena, as a consequence of which oxidative stress, decreased neurotransmitter level, elevated corticosterone level and altered NSC homeostasis is observed. However, the precise mechanism by which chronic stress induce anxious depression and modify internal milieu is still unknown. Herein, we show that exposure to CUS increase oxidative stress, microgliosis, astrogliosis while it reduces hippocampal NSC proliferation, neuronal differentiation and maturation in adult rats. CUS exposure in rats reduce dopamine and serotonin level in cortex and hippocampus, which result in increased anxiety and depression-like phenotypes. We also found elevated level of NF-κB and TNF-α while decreased anti-inflammatory cytokine IL-10 level, that led to increased expression of Bax and cleaved Caspase-3 whereas down regulation of antiapoptotic protein Bcl2. Additionally, CUS altered adult hippocampal neurogenesis, increased gliosis and neuronal apoptosis in cerebral cortex and hippocampus which might be associated with reduced AKT and increased ERK signaling, as seen in the rat brain tissue. Taken together, these results indicate that CUS induce oxidative stress and neuroinflammation which directly affects NSC dynamics, monoamines levels and behavioral functions in adult rats.

Introduction

The maintenance of ‘internal milieu’ inside living beings with changing environment is termed as “homeostasis”(Ahmad et al., 2010). Selye in 1956 reported that any disturbance in homeostasis is termed as “Stress”(Selye, 1956). Stressors are factors that cause stress and the response of living beings against these stressors is termed as “stress response”. However, stress response is an adaptive process which is mediated by endocrine, nervous and immune system (Chrousos, 2009). It has been shown that prolonged stress conditions may lead to tissue damage and degeneration in brain (Ahmad et al., 2010; Schneiderman et al., 2005). Hypothalamic paraventricular nucleus (PVN), anterior pituitary and adrenal glands are set mediators of stress response (Arnett et al., 2016), collectively named as hypothalamic- pituitary-adrenal (HPA) axis, which is mainly regulated by corticotrophin-releasing factor (CRF) (Rivier and Vale, 1983; Vale et al., 1981). Downstream effector of HPA axis is corticosterone, which exert its effect through glucocorticoid (GCs) receptors (Rivier and Vale, 1983; Vale et al., 1981). Clinical and pre-clinical studies have demonstrated that development and progression of various neurodegenerative diseases is associated with elevated GC levels (Conrad, 2008). Hippocampal neurodegeneration in stressed and corticosteroid-treated animals has been reported (Vyas et al., 2016). Previous reports suggest that oxidative stress plays a significant role in pathophysiology of depression, (Bajpai et al., 2014), which might be due to sensitivity of the central nervous system (CNS) to free radicals (Black et al., 2015). Chronic stress exert detrimental effect on several cell functions, including antioxidant system (Ahmad et al., 2010; Salim, 2017; Schiavone et al., 2013; Tripathi et al., 2017; Zafir and Banu, 2009). Oxidative stress mediated changes in neurotransmitter synthesis, degradation and alteration in protein homeostasis in brain are mainly responsible for neurodegenerative and psychiatric disorders (Kanarik et al., 2008; Lucca et al., 2009). Moreover, balanced synthesis of monoamines in CNS is essential for normal brain functioning (Flugge et al., 2004). Due to different type of stressors, change in monoamine level has been reported in various brain regions including nucleus accumbens, hippocampus and cortex (Joels and Baram, 2009). Brain is primary target and more vulnerable to oxidative damage due to high energy demand (Salim, 2017; Zafir and Banu, 2009). Majority of studies have reported increased oxidative burden during physiologically unfavorable conditions, thus causing damage to central monoaminergic system (Ahmad et al., 2010; Lilja, 1979). However, the proportion of oxidation products in monoaminergic system of brain, that can agitate redox state during stressful conditions, is not much known. Oxidation products of dopamine (DA) and serotonin (5-HT) are assumed to play a major role in neurodegenerative disease progression (Biosa et al., 2018). It has been suggested that cell- mediated immune activation and their long-term sequels are prime mediators of neurodegeneration and reduced adult neurogenesis, that characterize depression (Halliwell, 2006; Maes et al., 2009).

In brain, adult neurogenesis occurs via generation of functional neurons from adult neural precursor cells. It is a multistep process which continues throughout life, but only in restricted brain regions (Winner and Winkler, 2015). In adult brain two distinct regions generate new functional neurons; the dentate gyrus (DG) of hippocampus and the subventricular zone (SVZ) of lateral ventricles (Bond et al., 2015). SVZ provides a path for new born neurons to migrate from rostral migratory stream to the olfactory region (Sun et al., 2010). Adult neurogenesis also take place in other brain regions like neo cortex, striatum, amygdala and substantia nigra (Bond et al., 2015; Gould, 2007). Importantly, adult neurogenesis takes place in a gliogenic environment (Gotz et al., 2016). Unlike neural stem cells (NSCs) of embryonic stage, adult NSCs in DG of hippocampus are surrounded by many glial cell types, such as mature oligodendrocytes, microglia and astroglia cells (Kriegstein and Alvarez-Buylla, 2009). Adult neurogenesis requires a specific additional mechanism of neuronal fate specification and maintenance (Gotz et al., 2016; Grande et al., 2013; Herrera et al., 1999). Astrocytes and microglia are probably prime regulators of brain’s microenvironment and play both beneficial and detrimental roles (Block et al., 2006; Dohi et al., 2010). Microglia and astrocytes are extensively found in neurogenic niches, such as SVZ and SGZ (Mosher et al., 2012). In hippocampal neurogenesis, NPCs in SGZ of the DG give rise to new born neuroblasts, among these a small portion of neuroblasts join the hippocampal circuitry as mature neurons. The remaining newborn cells undergo apoptosis, which are further cleared through phagocytosis by microglia (Diaz-Aparicio et al., 2020; Sierra et al., 2010). Hence, microglia play an important role in adult hippocampal neurogenesis (Sierra et al., 2010). It has been seen that phagocytic property of microglia remains constant, although many of the newly generated neurons diminish with age and in other pathological conditions (Sierra et al., 2010). According to an earlier report, microglia and astrocytes have been shown to exert mixed effect on adult neurogenesis (Guttenplan and Liddelow, 2019). At the time of neuronal stress, initially these glial cells play protective role by producing various metabolites and neurotrophins to support neurons (Ransohoff and Perry, 2009; Sochocka et al., 2017; Xu et al., 2020). However, in case of chronic pathological condition, these functionally plastic glial cells undergo morphological changes, subsequently transformed from their basal latent stage to activated ramified form (Burda and Sofroniew, 2014; Jakel and Dimou, 2017; Rock et al., 2004) and secrete proinflammatory cytokines and free radicals (Ransohoff and Perry, 2009).

It has been reported that disease or injury induce hyperactivation of glial cells resulting in generation of ROS and RNS (Hu et al., 1995; Singh et al., 2019). Production of ROS and RNS cause malfunctioning of activated glial cells, which further result in lipid peroxidation and inflammation, that ultimately worsen neurological disorders (Jou, 2008; Lee et al., 1993). Inflammation is usually associated with innate immune system, which occurs in response to infection or injury, and leads to increased levels of proinflammatory cytokines, such as interleukin (IL)-1β, interleukin (IL)- 6, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ and decreased levels of anti-inflammatory cytokines (Zhang and An, 2007). Some of the evidences show the association of pathology of depression with inflammation of the peripheral and CNS (Fernandes et al., 2016; Miller and Raison, 2016). In CNS, cytokine network is made by neurons and glial cells, that produce cytokines and express cytokine receptors, additionally they also amplified cytokine signals, which has intense effect on neurotransmitter and corticotrophin releasing hormone (CRH) function, as well as on behavior (Dantzer, 2004; Raison et al., 2006; Raison and Miller, 2003). Earlier reports have shown presence of increased proinflammatory cytokines, chemokines, acute phase proteins, and cellular adhesion molecules in serum and cerebrospinal fluid (CSF) of depressed patients (Alesci et al., 2005; Maes, 1999; Sluzewska et al., 1995), while on the other hand, anti-inflammatory drugs as adjuvant therapy have shown positive effects in schizophrenia and depressive disorders (Al-Hakeim et al., 2015). Altered level of proinflammatory cytokines and chemokines in brain has been shown to affect neurotransmission, neuroendocrine activity and brain anatomy, which leads to neurotoxicity and induce emotional, cognitive, and behavioral changes (Haroon et al., 2012). Inflammatory and neurodegenerative hypothesis of depression show that enhanced neurodegeneration in depression is partly associated with chronic inflammatory response (Hurley and Tizabi, 2013; Maes et al., 2009). Neuroinflammation affects hypothalamus–pituitary– adrenal (HPA) axis and results in alteration of brain serotonin and adult neurogenesis in dentate gyrus of the hippocampus (Kim et al., 2016; Monje et al., 2011). Release of various neurotoxic cytokines, such as IL-6, IL1β, IL16, TNFα and IFN-γ etc. by pro-inflammatory microglial phenotypes and activated astrocytes has been shown to impair various stages of adult neurogenesis(Biscaro et al., 2012; Eddleston and Mucke, 1993; Wyss-Coray and Mucke, 2002), specifically causing reduction in NSC proliferation, neuronal differentiation and survival of adult newborn neurons (Choi et al., 2014; Iosif et al., 2006; Wu et al., 2012). One of the reports have shown reduced progenitor cell proliferation in IL-6 overexpressing transgenic mice(Vallieres et al., 2002). Although several studies have suggested that either endogenous or exogenous encounter of glucocorticoid in brain provide immunosuppressive effect (Kaye et al., 2000). However, in vivo evidences demonstrate that stress induced endogenous increase of glucocorticoids and exogenous glucocorticoid injection actually enhance immune function within the CNS. For example, increased proinflammatory cytokine level has been observed in rat brain and peripheral tissues after exposure to various stressors such as tail-shock stress, social isolation stress and immobilization stress in different experiments(Minami et al., 1991; Nguyen et al., 1998; Pugh et al., 1999). According to earlier reports, psychological and physical stress have potential to activate microglia in vivo through production of various inflammatory mediators, such as macrophage migration inhibitory factor (MIF), cyclooxygenase-2 (cox-2) nitric oxide, and 5-lipoxygenase (5-LO) (Madrigal et al., 2003; Niino et al., 2000; Uz et al., 1999). In addition to increase in inflammatory mediators and glucocorticoid level, chronic stress exposure cause excitotoxic damage that could be due to over-stimulation of the N-methyl-d-aspartate (NMDA) receptor and subsequent upsurge in extracellular glutamate, which ultimately results in dendritic atrophy, neuronal cell death and altered neurogenesis within the hippocampus (Stein-Behrens et al., 1994; Takahashi et al., 2002). Among above-described pathological conditions like microglia activation and diminished adult neurogenesis etc. due to chronic stress, some of these conditions are also seen in clinical data of MDD patients (Wang et al., 2018).

Emerging evidences demonstrate the involvement of Akt signaling in MDD (Beaulieu et al., 2009). An earlier clinical report has shown that phosphorylation of Akt decrease in hippocampus and PFC of suicide subjects (Dwivedi et al., 2010). ERK1/2 is considered as one of the leading signaling kinases associated with depression, an integrated transcriptome analysis has shown abnormalities in ERK1/2 signaling in PFC, derived from rat and humans, which demonstrate the crucial role of ERK1/2 signaling in vulnerability to develop depression (Malki et al., 2015). Oxidative stress induced neuronal apoptosis plays a key role in depression, according to previous reports, excessive free radicals mediated apoptosis reduce regional brain volume and neuronal cell body numbers (Shelton et al., 2011). Apoptosis is also reported in brain samples of suicidal subjects, where apoptotic outcomes are accompanied by a decrease in antiapoptotic and increase in proapoptotic mediators (Duman, 2009; Ilchibaeva et al., 2016; Kubera et al., 2011). Chronic stress has also been shown to dysregulate apoptosis in the DG (Lucassen et al., 2006; Pittenger and Duman, 2008). Many factors can influence hippocampal neurogenesis during adulthood (Balu and Lucki, 2009; Shohayeb et al., 2018). In this regard, few research articles have shown the deleterious effect of chronic exposure on both hippocampal neurogenesis and hippocampal-dependent behavior (Conrad, 2010; Joels et al., 2007; Mirescu and Gould, 2006). Chronic stress reduce proliferation, survival and neuronal differentiation of newly born cells (Joels et al., 2007; Koutmani and Karalis, 2015). Decreased hippocampal neurogenesis due to chronic stress markedly contributes to the mood related behavioral deficits in animals (Kempermann et al., 2008; Lagace et al., 2010; Pittenger and Duman, 2008).

Section snippets

Animals

Adult male Sprague Dawley rats (200−220 g) were used in the present study. Rats were housed 3 per cage in climate-controlled environment conditions, temperature (24 ± 2 °C) and 12/12 h. light/dark cycle (8:00 am to 8:00 pm) with ad libitum access to food and water. All animal protocols were approved by our Institutional Animals Ethics Committee (IAEC) ensuing the guidelines of CPCSEA (committee for the purpose of control and supervision of experiments on animals), in compliance with

CUS induce anxiety and depression-like behavior in adult rats

To evaluate the effect of CUS on anxiety and depression-like behavior along with social behavior and Anhedonia, we performed elevated plus test (EPM) (Fig. 1A, P < 0.01), and light and dark test (Fig. 1B, P < 0.01). We observed that CUS treated rats showed anxiety like behavior in EPM test, as they hesitated to spend more time in open arm of the test apparatus in comparison to the control group. EPM data showed that rats with CUS also displayed depression-like behavior as seen in light and dark

Discussion

Continual physical and psychological stress is known to be one of the causal factors for development of anxiety and major depression (Slavich and Irwin, 2014; Yang et al., 2015). Various researchers have found a link between oxidative stress and certain mood disorders (Bouayed et al., 2009; Salim, 2014). According to their findings, excessive levels of ROS disrupt neural cytoarchitecture and cause damage to biomolecules, such as lipids, nucleic acids and proteins (Chen et al., 2012; Fedoce et

CRediT authorship contribution statement

Parul designed and performed most of the experiments, Akanksha Mishra analyzed the data, Sonu Singh edited manuscript, Seema Singh conducted the Western blotting, Virendra Tiwari Conducted animal behaviour, Swati Chaturvedi and Muhammad Wahajuddin performed HPLC during study and Shubha Shukla conceived the Study, supervised and edited the manuscript

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgements

The authors would like to thank Director of CSIR-Central Drug Research Institute (CDRI), Lucknow, India for constant support during the study. The authors would like to thank Mrs. Sachi Bharti for her support in animal behavior related experiments. Parul, Akanksha Mishra, Seema Singh, Virendra Tiwari, Swati Chaturvedi are supported by a research fellowship from CSIR, New Delhi- India. Sonu Singh is supported by a research fellowship from Indian Council of Medical Research (ICMR), New Delhi-

References (172)

  • R. Dantzer

    Cytokine-induced sickness behaviour: a neuroimmune response to activation of innate immunity

    Eur. J. Pharmacol.

    (2004)
  • J.S. de Andrade et al.

    Chronic unpredictable mild stress alters an anxiety-related defensive response, Fos immunoreactivity and hippocampal adult neurogenesis

    Behav. Brain Res.

    (2013)
  • L. Diniz et al.

    Effects of chronic treatment with corticosterone and imipramine on fos immunoreactivity and adult hippocampal neurogenesis

    Behav. Brain Res.

    (2013)
  • Y. Dwivedi et al.

    Modulation in activation and expression of phosphatase and tensin homolog on chromosome ten, Akt1, and 3-phosphoinositide-dependent kinase 1: further evidence demonstrating altered phosphoinositide 3-kinase signaling in postmortem brain of suicide subjects

    Biol. Psychiatry

    (2010)
  • M. Eddleston et al.

    Molecular profile of reactive astrocytes--implications for their role in neurologic disease

    Neuroscience

    (1993)
  • M. Joels et al.

    Chronic stress: implications for neuronal morphology, function and neurogenesis

    Front. Neuroendocrinol.

    (2007)
  • M.J. Jou

    Pathophysiological and pharmacological implications of mitochondria-targeted reactive oxygen species generation in astrocytes

    Adv. Drug Deliv. Rev.

    (2008)
  • T.I. Kam et al.

    Microglia and astrocyte dysfunction in parkinson’s disease

    Neurobiol. Dis.

    (2020)
  • M. Kanarik et al.

    Changes in regional long-term oxidative metabolism induced by partial serotonergic denervation and chronic variable stress in rat brain

    Neurochem. Int.

    (2008)
  • R.J. Katz et al.

    Acute and chronic stress effects on open field activity in the rat: implications for a model of depression

    Neurosci. Biobehav. Rev.

    (1981)
  • Y.K. Kim et al.

    The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2016)
  • W.H. Kuang et al.

    IGF-1 defends against chronic-stress induced depression in rat models of chronic unpredictable mild stress through the PI3K/Akt/FoxO3a pathway

    Kaohsiung J. Med. Sci.

    (2018)
  • M. Kubera et al.

    In animal models, psychosocial stress-induced (neuro)inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2011)
  • S.C. Lee et al.

    Induction of nitric oxide synthase activity in human astrocytes by interleukin-1 beta and interferon-gamma

    J. Neuroimmunol.

    (1993)
  • S. Liang et al.

    Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress

    Neuroscience

    (2015)
  • K. Lingappan

    NF-kappaB in oxidative stress

    Curr. Opin. Toxicol.

    (2018)
  • G. Lucca et al.

    Effects of chronic mild stress on the oxidative parameters in the rat brain

    Neurochem. Int.

    (2009)
  • A. Ahmad et al.

    Alterations in monoamine levels and oxidative systems in frontal cortex, striatum, and hippocampus of the rat brain during chronic unpredictable stress

    Stress

    (2010)
  • S. Alesci et al.

    Major depression is associated with significant diurnal elevations in plasma interleukin-6 levels, a shift of its circadian rhythm, and loss of physiological complexity in its secretion: clinical implications

    J. Clin. Endocrinol. Metab.

    (2005)
  • M.G. Arnett et al.

    Genetic approaches to hypothalamic-pituitary-Adrenal Axis regulation

    Neuropsychopharmacology

    (2016)
  • D. Autheman et al.

    Glutathione peroxidase overexpression causes aberrant ERK activation in neonatal mouse cortex after hypoxic preconditioning

    Pediatr. Res.

    (2012)
  • A. Bajpai et al.

    Oxidative stress and major depression

    J. Clin. Diagn. Res.

    (2014)
  • J.M. Beaulieu et al.

    Akt/GSK3 signaling in the action of psychotropic drugs

    Annu. Rev. Pharmacol. Toxicol.

    (2009)
  • P. Belujon et al.

    Dopamine system dysregulation in major depressive disorders

    Int. J. Neuropsychopharmacol.

    (2017)
  • J.R. Bethea et al.

    Systemically administered interleukin-10 reduces tumor necrosis factor-alpha production and significantly improves functional recovery following traumatic spinal cord injury in rats

    J. Neurotrauma

    (1999)
  • A. Biosa et al.

    Dopamine oxidation products as mitochondrial endotoxins, a potential molecular mechanism for preferential neurodegeneration in parkinson’s disease

    ACS Chem. Neurosci.

    (2018)
  • B. Biscaro et al.

    Inhibition of microglial activation protects hippocampal neurogenesis and improves cognitive deficits in a transgenic mouse model for Alzheimer’s disease

    Neurodegener. Dis.

    (2012)
  • M.L. Block et al.

    Potent regulation of microglia-derived oxidative stress and dopaminergic neuron survival: substance P vs. Dynorphin

    FASEB J.

    (2006)
  • S.R. Bornstein et al.

    Approaching the shared biology of obesity and depression: the stress axis as the locus of gene-environment interactions

    Mol. Psychiatry

    (2006)
  • J. Bouayed et al.

    Oxidative stress and anxiety: relationship and cellular pathways

    Oxid. Med. Cell. Longev.

    (2009)
  • S. Cagnol et al.

    ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence

    FEBS J.

    (2010)
  • M.A. Calcia et al.

    Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness

    Psychopharmacology (Berl.)

    (2016)
  • C. Catale et al.

    Microglial function in the effects of early-life stress on brain and behavioral development

    J. Clin. Med.

    (2020)
  • S. Chakravarty et al.

    Chronic unpredictable stress (CUS)-induced anxiety and related mood disorders in a zebrafish model: altered brain proteome profile implicates mitochondrial dysfunction

    PLoS One

    (2013)
  • X. Chen et al.

    Oxidative stress in neurodegenerative diseases

    Neural Regen. Res.

    (2012)
  • S.S. Choi et al.

    Human astrocytes: secretome profiles of cytokines and chemokines

    PLoS One

    (2014)
  • G.P. Chrousos

    Stress and disorders of the stress system

    Nat. Rev. Endocrinol.

    (2009)
  • C.D. Conrad

    Chronic stress-induced hippocampal vulnerability: the glucocorticoid vulnerability hypothesis

    Rev. Neurosci.

    (2008)
  • J.N. Crawley

    Mouse behavioral assays relevant to the symptoms of autism

    Brain Pathol.

    (2007)
  • Z. Deng et al.

    Behavioral defects induced by chronic social defeat stress are protected by Momordica charantia polysaccharides via attenuation of JNK3/PI3K/AKT neuroinflammatory pathway

    Ann. Transl. Med.

    (2019)
  • Cited by (15)

    • Interleukin-1 beta in psychosocial stress

      2023, Stress: Immunology and Inflammation: Handbook of Stress Series Volume 5
    • Neuromodulation and hippocampal neurogenesis in depression: A scoping review

      2022, Brain Research Bulletin
      Citation Excerpt :

      Mutant mice with Bax deletion exhibited significantly increased neurogenesis, which prevented CORT-induced anxiety and depression-related behaviors (Hill et al., 2015). In CUMS/CUS models, it was reported that chronic stress promoted depressive-like behaviors and inhibited neurogenesis, which were accompanied by an upregulated apoptosis via increasing pro-apoptotic Bax and decreasing anti-apoptotic Bcl-2 in the hippocampus (Parul et al., 2021; Wang et al., 2014). Wang et al. showed that rTMS had an anti-apoptotic effect by normalizing the stress-induced changes in Bax and Bcl-2 expressions, therefore protected hippocampal neurons from apoptotic-induced cell death (Wang et al., 2014).

    View all citing articles on Scopus
    View full text