Elsevier

Experimental Neurology

Volume 295, September 2017, Pages 211-221
Experimental Neurology

Research Paper
Intranasal cotinine improves memory, and reduces depressive-like behavior, and GFAP + cells loss induced by restraint stress in mice

https://doi.org/10.1016/j.expneurol.2017.06.016Get rights and content

Highlights

  • Intranasal cotinine restored mood equilibrium after restraint stress in mice.

  • Intranasal cotinine recovered recognition memory after restraint stress in mice.

  • Intranasal cotinine recovered astrocytes morphology after restraint stress in mice.

Abstract

Posttraumatic stress disorder (PTSD), chronic psychological stress, and major depressive disorder have been found to be associated with a significant decrease in glial fibrillary acidic protein (GFAP) immunoreactivity in the hippocampus of rodents. Cotinine is an alkaloid that prevents memory impairment, depressive-like behavior and synaptic loss when co-administered during restraint stress, a model of PTSD and stress-induced depression, in mice. Here, we investigated the effects of post-treatment with intranasal cotinine on depressive- and anxiety-like behaviors, visual recognition memory as well as the number and morphology of GFAP + immunoreactive cells, in the hippocampus and frontal cortex of mice subjected to prolonged restraint stress. The results revealed that in addition to the mood and cognitive impairments, restraint stress induced a significant decrease in the number and arborization of GFAP + cells in the brain of mice. Intranasal cotinine prevented these stress-derived symptoms and the morphological abnormalities GFAP + cells in both of these brain regions which are critical to resilience to stress. The significance of these findings for the therapy of PTSD and depression is discussed.

Introduction

Although the effects of acute stress are usually brief and can be overcome quickly, exposure to chronic or extreme forms of unescapable stress can lead to neurochemical, morphological and functional changes in the brain that have been associated with posttraumatic stress disorder (PTSD) (North et al., 2016) and major depressive disorder (MDD) (Nestler et al., 2002). These stress-induced psychiatric diseases are characterized by symptoms such as intrusive distressing thoughts, anhedonia, irritability (McHugh et al., 2012), feelings of guilt, sleep disorders (Williams et al., 2015), cognitive impairment (Jak et al., 2016), anxiety (Wilder Schaaf et al., 2013) and sometimes treatment-resistant depression (Stander et al., 2014). Furthermore, these conditions have been associated with functional and structural changes in several regions of the brain including the amygdala (Laugharne et al., 2016), entorhinal cortex, prefrontal cortex and hippocampus (Meng et al., 2016, Sheynin and Liberzon, 2016, Yoon et al., 2017, Zhu et al., 2016). At cellular level, PTSD and depression are also associated with a decrease of glial fibrillary acidic protein (GFAP) immunoreactive cells (GFAP+) in the brain (Saur et al., 2016; Nestler et al., 2002; Fuller et al., 2010). GFAP is a family of proteins that includes eight isoforms expressed by different subpopulations of astrocytes as well as immature brain cells. These isoforms include GFAP+ 1, GFAP delta and GFAP kappa. GFAP delta appears to be linked with neural stem cells (NSCs) and may be involved in migration.

Furthermore, the expression of GFAP has been reported to decrease in response to microgravity (Day et al., 1998). Another predominantly-astroglial enzyme, glutamine synthase, has been reduced in the frontal cortex following intraventricular injection of aluminum (Guo-Ross et al., 1999), which paralleled alterations in GFAP expression. These results suggest an impairment of astrocytic responsivity in frontal cortex following toxic insults. Stress-induced depression is associated with a reduction of neurogenesis, neuroinflammation and a decrease of astrocytes in the brain (Sanacora and Banasr, 2013), and reduced GFAP expression has been found associated with schizophrenia, bipolar disorder and depression (Cobb et al., 2016, Webster et al., 2005). In addition, dysfunction of GFAP expression has been reported in encephalopathy (Kretzschmar et al., 1985). Astrocytes have been implicated in brain and neuronal functions supporting learning and memory and emotional responses (Dienel, 2017, Gibbs et al., 2006, Lee et al., 2014). Astrocytes have been identified as playing a significant role in maintaining brain homeostasis and supporting neuronal function (Weinstein et al., 1991). Coherent with these roles, it has been found a dysfunction of astroglia in the hippocampus of subjects with depression (Cobb et al., 2016) and other psychiatric and neurological conditions (Saur et al., 2016).

Because astrocytes support neuronal function and neurogenesis, it is thought that a decrease in astrocytic function is a critical factor underlying maladaptive responses in individuals with posttraumatic stress disorder (PTSD). Interestingly, treatment with antidepressants such as fluoxetine improved mood and induced a restoration of astroglia, suggesting that a restoration of astrocytes function is associated with changes in behavior.

An excellent model to investigate PTSD-induced, treatment-resistant depression (TRD) is the prolonged restraint stress paradigm (Perrine et al., 2016). Using this model, it has been shown that chronic stress induces cognitive deficits and depressive-like behavior, and morphological changes such as a decrease in the length and number of dendrites and synapses in neurons of the CA3 region of the hippocampus (Watanabe et al., 1992). Indeed, it has been shown that a two-hour immobilization stress combined with forced swim stress induced a decrease in the number of astrocytes in the hippocampus of rats (Imbe et al., 2012). Chronic restraint stress (6 h/day for 3 weeks), but not short-term restraint stress (6 h/day for 3 days), caused mechanical hypersensitivity and aggressive behavior. The chronic restraint stress induced a significant decrease of GFAP protein levels in the ventrolateral periaqueductal gray (Imbe et al., 2012). This decrease in astrocyte immunoreactivity (IR) was accompanied by a parallel decrease in glutamate transporter EAAT2 expression. The EAAT2 protein levels in the 3 week-stress group was significantly lower (− 20%) than in the control group. In contrast, there was no significant differences in the GFAP and EAAT2 protein levels between the three-day stress groups control in the periaqueductal gray matter (Imbe et al., 2012). It is thought that a deficit in glutamate transport after chronic stress may trigger neuronal dysfunction due to excitotoxicity in the brain.

Cotinine, a positive modulator of the α7 nicotinic acetylcholine receptors (nAChRs) (Echeverria et al., 2016b), has shown to decrease anxiety and to improve the extinction of fear in rodents subjected to fear conditioning a model of PTSD (de Aguiar et al., 2013, Zeitlin et al., 2012). Furthermore, continue treatment with oral cotinine prevented working memory loss and depressive-like behavior, as well as increased synaptic density in the hippocampus and frontal cortex of mice subjected to chronic immobilization stress (Grizzell et al., 2014a, Grizzell et al., 2014b). Cotinine has a long plasma half-life (19 to 24 h) and shows minor side effects in humans (Grizzell and Echeverria, 2015). Cotinine also has anti-inflammatory properties, overriding the production of cytokines that are under transcriptional control of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) system (transforming neurotrophic factor alpha (TNF-α), Interleukin (IL)-1β, IL-6, IL-12, IL-23) (Rehani et al., 2008).

The targets of cotinine are the most abundant nicotinic receptors in the brain, the low affinity (α7) and high affinity (α4β2) nAChRs (Colquhoun and Patrick, 1997). The activation of α7 receptors at the synapse triggers an increase in permeability to Na+ and Ca2 + ions (Pichon et al., 2004). These currents depolarize the cell and activate different neuronal signaling cascades (Exley and Cragg, 2008). Furthermore, Na+ and Ca2 + currents depolarize the pre-synaptic membrane, inducing the release of neurotransmitters such as dopamine, serotonin, glutamate, and gamma-amino butyric acid (GABA) (d'Incamps and Ascher, 2014, Exley and Cragg, 2008). In this way, the stimulation of the α7 and α4β2 receptors in the prefrontal cortex can promote the activation of glutamatergic neurons that normally inhibit the activity of the amygdala after chronic stress (Bencherif et al., 2014, Broide and Leslie, 1999). Thus, by acting on these receptors, cotinine may alleviate TRD in patients with PTSD.

Intranasal (IN) administration of CNS drug is acquiring increasing attention because of therapeutic advantages in reducing time of action of drugs, decreasing systemic effects and preventing hepatic first-pass elimination (Hanson and Frey, 2007, Hanson and Frey, 2008). In this study, in the search of new delivery methods for cotinine, we tested the behavioral effects of IN cotinine on depressive-like behavior, anxiety and working visual recognition memory when administered after prolonged restraint stress. In addition, to understand the mechanism of action of IN cotinine, we also examined its effect on the levels of GFAP+ cells in the hippocampus and frontal cortex of mice subjected or not to restraint stress. The significance of these results for the treatment of PTSD and TRD is discussed.

Section snippets

Animals

Mice were obtained from the animal facilities of the University of Chile, and maintained with free access to commercial food and water, in a controlled environment with an average temperature of 22 °C under a 12 h/12 h dark/light schedule. C57BL/6 male mice weighing between 20 and 30 grams (g) and aged about 2–3 months were used. Mice were acclimatized to the housing facility for a week before experiments. Test and animal care were performed according to protocols approved for the Universidad San

Statistical analysis

To analyze differences between-groups means in the behavioral and immunohistochemical studies, the following were used. Student's t-tests or Kruskal-Wallis were used when comparing two conditions and when comparing three or more levels of a factor, one-way followed by Tukey's or Tukey-Kramer post hoc tests (where applicable) or a repeated measure, 3 × 3 factorial ANOVA (treatment condition × brain region) followed by Fisher's LSD post-hoc tests were used where appropriate. For the IHC analyses of

Effect of posttreatment with intranasal cotinine on stress-induced changes in locomotor activity

To assess changes in locomotor activity related to restraint stress and cotinine treatment, we first tested each mouse in the open field, a task that permits investigators to assess changes in locomotor activity and anxiety behavior. One-way ANOVA analysis revealed significant differences between treatment groups in locomotor activity, expressed as distance travelled in the OF test (F (2,17) = 5.144, p = 0.018). A Tukey Post-hoc analysis indicated a significant increase (p < 0.05) in locomotor

Discussion

Chronic stress in rodents is considered a good animal model to investigate antidepressants for treatment-resistant depression (TRD) in PTSD. In this work, the effects of post-treatment with IN cotinine on behavior and GFAP + cells in the hippocampus and frontal cortex of adult male mice subjected to stress were investigated. The results show that IN cotinine normalized the otherwise abnormal behavior in the chronically stressed mice. In addition, we found a clear effect of intranasal cotinine on

Conclusions

The evidence obtained in this study permits to conclude that post-treatment with IN cotinine is effective in restoring mood equilibrium and cognitive abilities as well as astrocytes function after chronic restraint stress in mice. The preceding constitutes the first evidence about the action of cotinine on GFAP+ cells. This finding represents a new mechanism of action of cotinine to restore neuronal survival and plasticity after stress. The IN delivery of cotinine proved to be effective as a

Acknowledgements

This material is the result of work supported with resources and the use of facilities at the Bay Pines VA Healthcare System. The contents do not necessarily represent the views of the Department of Veterans Affairs or the United States Government. Also, we thank Dr. William Fray and Dr. Leha Hanson from the Alzheimer's Research Center at Regions Hospital, HealthPartners Research Foundation for providing training and guidance with the implementation of intranasal delivery of drugs to mice. We

References (102)

  • D. Forster et al.

    Nucleotides protect rat brain astrocytes against hydrogen peroxide toxicity and induce antioxidant defense via P2Y receptors

    Neurochem. Int.

    (2016)
  • S. Fuller et al.

    Activated astroglia during chronic inflammation in Alzheimer's disease – do they neglect their neurosupportive roles?

    Mutat. Res.

    (2010)
  • J. Gao et al.

    Evaluation of nicotine and cotinine analogs as potential neuroprotective agents for Alzheimer's disease

    Bioorg. Med. Chem. Lett.

    (2014)
  • B. Grayson et al.

    Assessment of disease-related cognitive impairments using the novel object recognition (NOR) task in rodents

    Behav. Brain Res.

    (2015)
  • J.A. Grizzell et al.

    Cotinine reduces depressive-like behavior, working memory deficits, and synaptic loss associated with chronic stress in mice

    Behav. Brain Res.

    (2014)
  • J.A. Grizzell et al.

    Cotinine improves visual recognition memory and decreases cortical Tau phosphorylation in the Tg6799 mice

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2017)
  • S.C. Heinrichs et al.

    Repeated valproate treatment facilitates fear extinction under specific stimulus conditions

    Neurosci. Lett.

    (2013)
  • H. Imbe et al.

    Chronic restraint stress decreases glial fibrillary acidic protein and glutamate transporter in the periaqueductal gray matter

    Neuroscience

    (2012)
  • T. Karl et al.

    Behavioral phenotyping of mice in pharmacological and toxicological research

    Exp. Toxicol. Pathol.

    (2003)
  • Y. Liu et al.

    Activation of alpha7 nicotinic acetylcholine receptors protects astrocytes against oxidative stress-induced apoptosis: implications for Parkinson's disease

    Neuropharmacology

    (2015)
  • T. McHugh et al.

    Anger in PTSD: is there a need for a concept of PTSD-related posttraumatic anger?

    Clin. Psychol. Rev.

    (2012)
  • E.J. Nestler et al.

    Neurobiology of depression

    Neuron

    (2002)
  • M.A. Orlovsky et al.

    Hippocampus remodeling by chronic stress accompanied by GR, proteasome and caspase-3 overexpression

    Brain Res.

    (2014)
  • N. Otani et al.

    Enhanced hippocampal neurodegeneration after traumatic or kainate excitotoxicity in GFAP-null mice

    J. Clin. Neurosci.

    (2006)
  • S.A. Perrine et al.

    Severe, multimodal stress exposure induces PTSD-like characteristics in a mouse model of single prolonged stress

    Behav. Brain Res.

    (2016)
  • K. Rehani et al.

    Cotinine-induced convergence of the cholinergic and PI3 kinase-dependent anti-inflammatory pathways in innate immune cells

    Biochim. Biophys. Acta

    (2008)
  • G. Sanacora et al.

    From pathophysiology to novel antidepressant drugs: glial contributions to the pathology and treatment of mood disorders

    Biol. Psychiatry

    (2013)
  • A.E. Schaffner et al.

    The effect of type 1 astrocytes on neuronal complexity: a fractal analysis

    Methods

    (2001)
  • V.A. Stander et al.

    Etiology of depression comorbidity in combat-related PTSD: a review of the literature

    Clin. Psychol. Rev.

    (2014)
  • J. Szczepanik et al.

    Amygdala response to explicit sad face stimuli at baseline predicts antidepressant treatment response to scopolamine in major depressive disorder

    Psychiatry Res.

    (2016)
  • Y. Watanabe et al.

    Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons

    Brain Res.

    (1992)
  • M.J. Webster et al.

    Glial fibrillary acidic protein mRNA levels in the cingulate cortex of individuals with depression, bipolar disorder and schizophrenia

    Neuroscience

    (2005)
  • K.M. Wildeboer-Andrud et al.

    Cotinine impacts sensory processing in DBA/2 mice through changes in the conditioning amplitude

    Pharmacol. Biochem. Behav.

    (2014)
  • K.P. Wilder Schaaf et al.

    Anxiety, depression, and PTSD following cardiac arrest: a systematic review of the literature

    Resuscitation

    (2013)
  • L. Xia et al.

    FGF2 blocks PTSD symptoms via an astrocyte-based mechanism

    Behav. Brain Res.

    (2013)
  • S.S. Yang et al.

    Effects of SPAK knockout on sensorimotor gating, novelty exploration, and brain area-dependent expressions of NKCC1 and KCC2 in a mouse model of schizophrenia

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2015)
  • R.B. de Aguiar et al.

    Neuroactive effects of cotinine on the hippocampus: behavioral and biochemical parameters

    Neuropharmacology

    (2013)
  • M. Antunes et al.

    The novel object recognition memory: neurobiology, test procedure, and its modifications

    Cogn. Process.

    (2012)
  • T.L. Babb et al.

    Astrocytes may contribute to the latent period in progressive neuron loss, axon sprouting, and chronic seizures in rat kainate hippocampal epilepsy

    Epilepsy Res. Suppl.

    (1996)
  • G.E. Barreto et al.

    Nicotine-derived compounds as therapeutic tools against post-traumatic stress disorder

    Curr. Pharm. Des.

    (2015)
  • G.E. Barreto et al.

    Advances in medicinal plants with effects on anxiety behavior associated to mental and Health conditions

    Curr. Med. Chem.

    (2017)
  • M. Bencherif et al.

    Alpha7 neuronal nicotinic receptor: a pluripotent target for diseases of the central nervous system

    CNS Neurol. Disord. Drug Targets

    (2014)
  • R.S. Broide et al.

    The alpha7 nicotinic acetylcholine receptor in neuronal plasticity

    Mol. Neurobiol.

    (1999)
  • B. Czeh et al.

    Astroglial plasticity in the hippocampus is affected by chronic psychosocial stress and concomitant fluoxetine treatment

    Neuropsychopharmacology

    (2006)
  • B. Czeh et al.

    Chronic social stress inhibits cell proliferation in the adult medial prefrontal cortex: hemispheric asymmetry and reversal by fluoxetine treatment

    Neuropsychopharmacology

    (2007)
  • J.R. Day et al.

    Effects of microgravity and bone morphogenetic protein II on GFAP in rat brain

    J. Appl. Physiol.

    (1998)
  • D.G. Diniz et al.

    Age, environment, object recognition and morphological diversity of GFAP-immunolabeled astrocytes

    Behav. Brain Funct.

    (2016)
  • V. Echeverria et al.

    Neuroinflammation: a therapeutic target of cotinine for the treatment of psychiatric disorders?

    Curr. Pharm. Des.

    (2016)
  • R. Exley et al.

    Presynaptic nicotinic receptors: a dynamic and diverse cholinergic filter of striatal dopamine neurotransmission

    Br. J. Pharmacol.

    (2008)
  • K.B.J. Franklin et al.

    The Mouse Brain in Stereotaxic Coordinates

    (2001)
  • Cited by (22)

    • Traditional herbal formula Jiao-tai-wan improves chronic restrain stress-induced depression-like behaviors in mice

      2022, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      They can release a variety of signaling molecules including adenosine, adenosine ATP and glutamate, which are involved in the regulation of depressive-like behaviors[125,126]. Studies have shown that the number of GFAP+ cells were decreased in the hippocampus and medial prefrontal cortex MDD patients and chronic mild stress-induced rodent models[127–129]. However, the changes of astrocytes in other neurological diseases such as Parkinson's disease, Alzheimer's disease and stroke are different from those in MDD patients, which may be due to the fact that in addition to astrocytes, there are other factors involved in the pathogenesis of these diseases[130–134].

    • Leukemia Inhibitory Factor Participates in the Formation of Stress Adaptation via Hippocampal Myelination in Mice

      2020, Neuroscience
      Citation Excerpt :

      Furthermore, 5-HT1A receptors have been reported to be expressed in astrocytes, and play a significant role in their proliferation (Miyazaki et al., 2013). Astrocytes have been implicated in brain and neuronal function supporting learning, memory and emotional responses (Lee et al., 2014; Diniz et al., 2016; Perez-Urrutia et al., 2017). A previous study reported that restraint stress for 6 h/day for 21 days induced depressive-like behaviors, and decreased the number and arborization of GFAP+ cells in the dentate gyrus of the hippocampus (Perez-Urrutia et al., 2017).

    • Effects of chronic mild stress induced depression on synaptic plasticity in mouse hippocampus

      2019, Behavioural Brain Research
      Citation Excerpt :

      Neurotransmitter mediated Ca2+ increase of astrocyte which may lead to glutamate release that modulate neuronal activity and synaptic transmission [10], the astrocyte function loss may result from activated HPA axis with increased glucocorticoid and corticosterone which related to neuroinflammation [45]. Furthermore, increased glucocorticoid and corticosterone could lead to apoptosis and autophagy of neurons and glia by activating glutamate in depression [46–48], major depressive disorder is associated with the decrease of GFAP in the hippocampus [49] and apoptosis of astrocytes in PFC [50] of rodents. In consistent with previous reports, we found a downregulated level of GFAP as well, which probably caused by apoptosis of astrocytes.

    • Cotinine and memory: Remembering to forget

      2019, Neuroscience of Nicotine: Mechanisms and Treatment
    • Deciphering sex differences in the immune system and depression

      2018, Frontiers in Neuroendocrinology
      Citation Excerpt :

      Johnson et al. (2008) determined that functional androgen receptors (ARs) are required for the masculinization of astrocytes in the medial amygdala. Animal and human postmortem studies suggest astrocytes are involved in major depressive disorder (Altshuler et al., 2010; Gosselin et al., 2009; Perez-Urrutia et al., 2017; Rajkowska and Stockmeier, 2013). Altshuler et al. (2010) examined postmortem amygdala samples from subjects diagnosed with bipolar disorder, major depressive disorder, and schizophrenia, as well as controls.

    View all citing articles on Scopus
    View full text