Elsevier

Brain, Behavior, and Immunity

Volume 69, March 2018, Pages 556-567
Brain, Behavior, and Immunity

Full-length Article
TNFα disrupts blood brain barrier integrity to maintain prolonged depressive-like behavior in mice

https://doi.org/10.1016/j.bbi.2018.02.003Get rights and content

Highlights

  • We describe a prolonged (>4 weeks) learned helplessness (LH) depression model in mice.

  • Prolonged LH was associated with elevated hippocampal TNFα, IL-17A and IL-23 levels.

  • Blood-brain barrier (BBB) disruption was evident in mice with prolonged LH.

  • Prolonged LH and elevated cytokines were rapidly reversed by Fingolimod or TNFα inhibitor treatment.

  • TNFα-mediated disruption of the BBB may contribute to impaired recovery from LH.

Abstract

Recovery from major depressive disorder is difficult, particularly in patients who are refractory to antidepressant treatments. To examine factors that regulate recovery, we developed a prolonged learned helplessness depression model in mice. After the induction of learned helplessness, mice were separated into groups that recovered or did not recover within 4 weeks. Comparisons were made between groups in hippocampal proteins, inflammatory cytokines, and blood brain barrier (BBB) permeability. Compared with mice that recovered and control mice, non-recovered mice displaying prolonged learned helplessness had greater hippocampal activation of glycogen synthase kinase-3 (GSK3), higher levels of tumor necrosis factor-α (TNFα), interleukin-17A, and interleukin-23, increased permeability of the blood brain barrier (BBB), and lower levels of the BBB tight junction proteins occludin, ZO1, and claudin-5. Treatment with the GSK3 inhibitor TDZD-8 reduced inflammatory cytokine levels, increased tight junction protein levels, and reversed impaired recovery from learned helplessness, demonstrating that prolonged learned helplessness is reversible and is maintained by abnormally active GSK3. In non-recovered mice with prolonged learned helpless, stimulation of sphingosine 1-phosphate receptors by Fingolimod or administration of the TNFα inhibitor etanercept repaired the BBB and reversed impaired recovery from prolonged learned helplessness. Thus, disrupted BBB integrity mediated in part by TNFα contributes to blocking recovery from prolonged learned helplessness depression-like behavior. Overall, this report describes a new model of prolonged depression-like behavior and demonstrates that stress-induced GSK3 activation contributes to disruption of BBB integrity mediated by inflammation, particularly TNFα, which contributes to impaired recovery from prolonged learned helplessness.

Introduction

Major depressive disorder, which afflicts nearly 17% of people in the United States, is defined by the presence of multiple debilitating symptoms and by their prolonged duration (Kessler et al., 2005). Stress is a common precipitating factor of depression, and stress is often used to induce rodent models of depression (Kessler, 1997, Pariante and Lightman, 2008). These rodent models are evaluated by depression-like behaviors because rodents are not thought to experience the equivalent of major depressive disorder (Gould and Gottesman, 2006, Nestler and Hyman, 2010).

One robust rodent depression model is learned helplessness, in which rodents are exposed to uncontrollable and inescapable mild foot shocks as the aversive stimuli (Chourbaji et al., 2005, Maier and Seligman, 2016, Vollmayr and Gass, 2013). Upon subsequent exposure to foot shocks with escape freely available, rodents that display learned helplessness fail to leave the foot shock chamber. Learned helplessness is considered a behavioral model of depression, particularly involving despair and a coping deficit in response to stress, because it is accompanied by changes typical of depression, including decreased brain regional levels of serotonin, norepinephrine, dopamine, and brain-derived neurotrophic factor (BDNF), decreased home cage activity, elevated corticosteroid levels, increased rapid eye movement during sleep, and displays of other depression-like behaviors, such as impaired social interactions, anhedonia, and increased immobility in the tail suspension and forced swim tests (Chourbaji et al., 2005, Henn and Vollmayr, 2005, Vollmayr and Gass, 2013). Furthermore, susceptibility to learned helplessness is diminished by treatment with antidepressants and other drugs, such as a high dose of lithium (Beurel et al., 2011, Vollmayr and Gass, 2013). Lithium is an inhibitor of glycogen synthase kinase-3 (GSK3), which refers to two isoforms that are predominantly controlled by inhibitory phosphorylation on serine21-GSK3α and serine9-GSK3β (Beurel et al., 2015). Substantial evidence indicates that abnormally active GSK3 promotes depression and animal models of depression, which may derive in part from the promotion of inflammation by GSK3 (Jope, 2011).

Numerous studies have reported treatments and mechanisms that regulate susceptibility to onset of depression-like behaviors in rodents (Gould and Gottesman, 2006, Nestler and Hyman, 2010), but less is known about treatments and mechanisms that control recovery from prolonged depression-like behaviors. We observed that some mice display prolonged learned helplessness, rather than recovering in the usual 2–4 week period. This led us to examine mechanisms that may contribute to impaired recovery. Inflammatory cytokines are elevated in depressed patients, including treatment refractory patients. One of the possible mechanisms whereby cytokines can maintain depressive symptoms is by affecting the blood brain barrier (BBB). There is evidence of impaired integrity of the BBB in depressed patients (Lavoie et al., 2010, Najjar et al., 2013) and rodent models of depression (Esposito et al., 2001, Friedman et al., 1996, Santha et al., 2015, Sharma et al., 1995), and inflammatory cytokines, such as tumor necrosis factor-α (TNFα), can impair BBB integrity (Rochfort and Cummins, 2015). Endothelial cells connected by tight junctions (TJs) constitute an integral component of the BBB. TJs are comprised of occludin, claudin, ZO-1, ZO-2 and other junction adhesion molecules that maintain structural and functional integrity of the brain endothelium (Obermeier et al., 2013). GSK3 inhibition promotes TJ stability in brain endothelial cells by increasing the levels of occludin and claudin-5 (Ramirez et al., 2013), suggesting GSK3 inhibition may protect the BBB by stabilizing TJ proteins. Decreased levels of TJ proteins and BBB breakdown occur after trauma, stroke, multiple sclerosis, and Alzheimer’s disease, conditions associated with increased activated GSK3, neuroinflammation and increased prevalence of depression.

Therefore, we examined links between recovery from prolonged learned helplessness and GSK3, inflammation and BBB integrity. The results show that BBB permeability is increased in mice that do not recover from prolonged learned helplessness, and that recovery and BBB integrity can be improved pharmacologically by treatment with a GSK3 inhibitor, Fingolimod, or anti-TNFα. These results indicate that stress-induced GSK3 activation promotes inflammation that contributes to disruption of BBB integrity, and this is associated with impaired recovery from prolonged learned helplessness.

Section snippets

Mice and drug administration

These studies used 8–12 week old male wild-type C57BL/6 mice from the National Cancer Institute and Charles River Laboratory. All mice were housed and treated in accordance with NIH and the University of Miami Institutional Animal Care and Use Committee regulations. Mice were treated TDZD-8 (5mg/kg; produced in the Martinez laboratory (Martinez et al., 2002)) which directly inhibits both isoforms of GSK3, the sphingosine 1-phosphate receptor (S1PR) agonist Fingolimod (FTY-720; 1 mg/kg;

Mice unable to recover from learned helplessness display increased active hippocampal GSK3

After inducing learned helplessness, mice were retested with escapable foot shocks once a week to determine the rate of recovery. Whereas ∼80% of mice recovered from learned helplessness within 3–4 weeks (Fig. 1A), after 4 and 5 weeks ∼20% of mice remained learned helpless (hereafter referred to as non-recovered mice). The mice that remained learned helpless for 5 weeks had 29 ± 0.3, 29 ± 0.5, 28 ± 0.7, 28 ± 0.7 and 27 ± 0.9 escape failures in weeks 1, 2, 3, 4, and 5, respectively,

Discussion

We describe here a mouse model for studying mechanisms that regulate recovery from a long-term depression-like behavior, which revealed that disrupted BBB integrity is associated with impaired recovery from prolonged learned helplessness. In this model, ∼20% of mice maintained learned helplessness for at least 5 weeks, whereas ∼80% spontaneously recovered within 1–4 weeks. This model differs from others that apply the inducing stressors repeatedly to maintain depression, such as chronic

Acknowledgments

This research was supported by grants from the NIMH (MH104656, MH110415, MH095380), a Merit Award from the Veterans Administration (BX003678), and a NARSAD Distinguished Investigator Grant from the Brain & Behavior Research Foundation. We thank Dr. Paul Smith, Novartis, for the gift of Fingolimod.

References (59)

  • F.A. Henn et al.

    Stress models of depression: forming genetically vulnerable strains

    Neurosci. Biobehav. Rev.

    (2005)
  • U. Krugel et al.

    Antidepressant effects of TNF-alpha blockade in an animal model of depression

    J. Psychiatr. Res.

    (2013)
  • J.F. Lee et al.

    Dual roles of tight junction-associated protein, zonula occludens-1, in sphingosine 1-phosphate-mediated endothelial chemotaxis and barrier integrity

    J. Biol. Chem.

    (2006)
  • B.S. McKenzie et al.

    Understanding the IL-23-IL-17 immune pathway

    Trends Immunol.

    (2006)
  • V.E. Miron et al.

    Central nervous system-directed effects of FTY720 (fingolimod)

    J. Neurol. Sci.

    (2008)
  • C.M. Pariante et al.

    The HPA axis in major depression: classical theories and new developments

    Trends Neurosci.

    (2008)
  • B. Prager et al.

    Sphingosine 1-phosphate signaling at the blood-brain barrier

    Trends Mol. Med.

    (2015)
  • G.A. Rosenberg et al.

    Tumor necrosis factor-alpha-induced gelatinase B causes delayed opening of the blood-brain barrier: an expanded therapeutic window

    Brain Res.

    (1995)
  • H.S. Sharma et al.

    Probable involvement of serotonin in the increased permeability of the blood-brain barrier by forced swimming. An experimental study using Evans blue and 131I-sodium tracers in the rat

    Behav. Brain Res.

    (1995)
  • B.B. Simen et al.

    TNFalpha signaling in depression and anxiety: behavioral consequences of individual receptor targeting

    Biol. Psychiatry

    (2006)
  • C. Zhao et al.

    TNF-Alpha knockout and minocycline treatment attenuates blood brain barrier leakage in MPTP-treated mice

    Neurobiol. Dis.

    (2007)
  • E. Beurel et al.

    Inhibition of glycogen synthase kinase-3 is necessary for the rapid antidepressant effect of ketamine in mice

    Mol. Psychiatry

    (2011)
  • S. Campbell et al.

    The role of the hippocampus in the pathophysiology of major depression

    J. Psychiatry Neurosci.

    (2004)
  • L. di Nuzzo et al.

    Antidepressant activity of fingolimod in mice

    Pharmacol. Res. Perspect.

    (2015)
  • C.A. Foster et al.

    FTY720 rescue therapy in the dark agouti rat model of experimental autoimmune encephalomyelitis: expression of central nervous system genes and reversal of blood-brain-barrier damage

    Brain Pathol.

    (2009)
  • A. Friedman et al.

    Pyridostigmine brain penetration under stress enhances neuronal excitability and induces early immediate transcriptional response

    Nat. Med.

    (1996)
  • J.G. Garcia et al.

    Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement

    J. Clin. Invest.

    (2001)
  • S.A. Golden et al.

    Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression

    Nat. Med.

    (2013)
  • T.D. Gould et al.

    Psychiatric endophenotypes and the development of valid animal models

    Genes Brain Behav.

    (2006)
  • Cited by (164)

    View all citing articles on Scopus
    1

    These authors contributed equally to this research.

    View full text