Elsevier

Brain, Behavior, and Immunity

Volume 41, October 2014, Pages 65-81
Brain, Behavior, and Immunity

Neuropathic pain-induced depressive-like behavior and hippocampal neurogenesis and plasticity are dependent on TNFR1 signaling

https://doi.org/10.1016/j.bbi.2014.04.003Get rights and content

Abstract

Patients suffering from neuropathic pain have a higher incidence of mood disorders such as depression. Increased expression of tumor necrosis factor (TNF) has been reported in neuropathic pain and depressive-like conditions and most of the pro-inflammatory effects of TNF are mediated by the TNF receptor 1 (TNFR1). Here we sought to investigate: (1) the occurrence of depressive-like behavior in chronic neuropathic pain and the associated forms of hippocampal plasticity, and (2) the involvement of TNFR1-mediated TNF signaling as a possible regulator of such events. Neuropathic pain was induced by chronic constriction injury of the sciatic nerve in wild-type and TNFR1−/− mice. Anhedonia, weight loss and physical state were measured as symptoms of depression. Hippocampal neurogenesis, neuroplasticity, myelin remodeling and TNF/TNFRs expression were analyzed by immunohistochemical analysis and western blot assay.

We found that neuropathic pain resulted in the development of depressive symptoms in a time dependent manner and was associated with profound hippocampal alterations such as impaired neurogenesis, reduced expression of neuroplasticity markers and myelin proteins. The onset of depressive-like behavior also coincided with increased hippocampal levels of TNF, and decreased expression of TNF receptor 2 (TNFR2), which were all fully restored after mice spontaneously recovered from pain. Notably, TNFR1−/− mice did not develop depressive-like symptoms after injury, nor were there changes in hippocampal neurogenesis and plasticity.

Our data show that neuropathic pain induces a cluster of depressive-like symptoms and profound hippocampal plasticity that are dependent on TNF signaling through TNFR1.

Introduction

Over half of all patients who suffer from neuropathic pain develop mood disorders such as depression and anxiety (Maletic and Raison, 2009, McWilliams et al., 2003), but the mechanisms underlying this comorbidity are not fully understood. Accumulating evidence suggests a role for the immune system in the etiology of depression (Eyre and Baune, 2012). Elevated levels of immune mediators such as TNF, have been detected in depressed patients (Mikova et al., 2001, Tuglu et al., 2003), while in rodents high levels of cytokines induce a depressive-like behavior, known as “sickness behavior” (Hart, 1988, Kaster et al., 2012). This condition can be reliably reproduced with the administration of cytokines or cytokine-inducers (Harrison et al., 2009, Yirmiya, 1996), and blocked by cytokine antagonists, or anti-inflammatory cytokines (Dantzer, 2001, Kent et al., 1992, Shamash et al., 2002). Moreover, genetically modified mice that do not express TNF receptors (TNFRs) are more resistant to the development of depressive behavior under stressful conditions, while TNF administration renders mice more susceptible to depression (Simen et al., 2006). It has been shown that antidepressants can reduce plasma TNF concentration (Kubera et al., 2005, Yirmiya et al., 1999), and in clinical trials, in which TNFRs antagonists were used for the treatment of immune pathologies, a significant improvement of depressive symptoms was observed (Bos and de Korte, 2006, Ertenli et al., 2012, Tyring et al., 2006).

TNF signals via 2 distinct receptors which often mediate opposing biological functions: the pro-inflammatory/pro-neurodegenerative/pro-demyelinanting TNF receptor 1 (TNFR1/p55) and the likely neuroprotective TNF receptor 2 (TNFR2/p75) (Baud and Karin, 2001, Brambilla et al., 2011, MacEwan, 2002). Interestingly, TNF has been proven to have a key role in the development of neuropathic pain (George et al., 2004, Martuscello et al., 2012), which has been associated to its action through TNFR1 (Schafers et al., 2002, Vogel et al., 2006).

The hippocampus, a central component of the limbic system, is a crucial mood-regulating region of the brain, also involved in the processing of nociception (Mutso et al., 2012). With the discovery of new neuron formation in this area of the adult brain, significant emphasis has been ascribed to the role of the neurogenic process in mood regulation and impairment of adult hippocampal neurogenesis has been linked to the development of depression (Sahay and Hen, 2007). However, other neuroplastic changes such as reduced spine density and dendritic retraction, were previously shown to occur at this level in animal models of depression or pain (Duman and Charney, 1999, Kodama et al., 2007, Watanabe et al., 1992) and, as with the neurogenic process, these alterations can be reverted by treatment with antidepressants as animals recover from depressive-like symptoms (Reinés et al., 2008, Warner-Schmidt and Duman, 2006). It is noteworthy that impairments in brain white matter have been described in psychiatric diseases such as schizophrenia and depression (Kyriakopoulos et al., 2009, Mettenburg et al., 2012), and specifically have been found to be associated with the limbic system in the melancholic subtype of major depressive disorder (Korgaonkar et al., 2011). Interestingly, Zeng et al. (2012) showed that white matter volume is normalized by antidepressant treatment in patients with major depression. So far little is known about the mechanisms implicated in white matter impairments that occurs in depressed patients. Conversely, demyelinating disorders characterized by myelin loss show co-morbidity with depression (Arnett et al., 2008), yet, the possible contribution of myelin remodeling as part of the hippocampal plasticity that occurs in depression has not been addressed.

Notably, it has been shown that TNF can be detrimental for the survival of the new hippocampal neurons (Cacci et al., 2005, Monje et al., 2003) and that TNF receptors have a differential actions in the modulation of hippocampal neurogenesis; TNFR1 acts as a suppressor of adult neurogenesis, whereas the absence TNFR2 results in reduced hippocampal neurogenesis (Chen and Palmer, 2013, Iosif et al., 2006). Interestingly, TNF has been shown to have a role in the pathophysiology of autoimmune demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) (Finsen et al., 2002) and the role of TNFR1-mediated TNF signaling in causing demyelination has been widely investigated by different authors (Arnett et al., 2001, Eugster et al., 1999, Probert et al., 2000). Finally, we and others have demonstrated that signaling mediated by transmembrane TNF, occurring mainly through TNFR2, has been proven to be essential for axon and myelin preservation and to promote remyelination (Bracchi-Ricard et al., 2013, Brambilla et al., 2011, Taoufik et al., 2011).

Based upon this evidence we sought to determine whether neuropathic pain-associated depression is related to hippocampal neuroplasticity and myelin alterations, and whether TNF signaling through TNFR1 plays a role in these events. We hypothesized that chronic neuropathic pain would be associated with the occurrence of depressive-like symptoms and that this would be mediated by hippocampal TNF/TNFR1 signaling resulting in impairment of hippocampal neurogenesis and neuroplasticity. Given the role of TNF/TNFR1 in demyelinating pathologies we also tested the hypothesis of myelin remodeling occurring in the hippocampus of mice suffering from neuropathic-pain-induced depression. To this purpose we used wild-type and TNFR1−/− mice to investigate: (1) the occurrence of depressive-like symptoms, such as anhedonia, loss of body weight gain and self-neglect in mice with neuropathic pain; (2) hippocampal neurogenesis, neuronal plasticity and myelin remodeling and (3) hippocampal TNF and TNFRs expression at the onset and recovery from neuropathic pain.

Our data show a temporal relationship between neuropathic pain and the occurrence of depressive-like symptoms as well as structural neuroplastic and white matter impairments of the hippocampus. Moreover, this is the first report to show that neuropathic pain-induced depression is associated with hippocampal TNF activity through TNFR1 and that this correlates with profound hippocampal plasticity.

Section snippets

Animals

TNFR1 null mice (TNFR1 KO) carrying the Tnfrsf1atm1Mak mutation (TNFR1−/−; Pfeffer et al., 1993), were purchased from Jackson Laboratory and backcrossed to C57BL/6 genetic background by breeding with C57BL/6 wild-type (WT) mice (Jackson Laboratory) for 9 generations. Resulting heterozygous mice were then interbred to produce TNFR1−/− mice and wild-type littermate controls for experiments. The genotypes were confirmed by polymerase chain reaction (PCR) from tail DNA (Supplementary 1A). The

CCI-induced neuropathic pain symptoms precede and parallel the development and resolution of depressive like-behavior

Mice subjected to CCI of the right sciatic nerve developed mechanical allodynia and thermal hyperalgesia on the ipsilateral hind paw, but not on the controlateral paw (two-way ANOVA, injured side vs uninjured side, allodynia: F1,924 = 1415.61, p < 0.0001; hyperalgesia: F1,726 = 205.89, p < 0.0001; CCI, n = 34, sham, n = 26). In particular mice developed mechanical allodynia one week after surgery (0.2 ± 0.04 g injured side vs 1.0 ± 0.02 g uninjured side, p < 0.001), and were completely recovered 12 weeks later (1.0 ±

Discussion

Our data show a temporal relationship between CCI-induced neuropathic pain and the occurrence of depressive behavior, as well as decreased neurogenesis, neuroplasticity and myelin remodeling in the hippocampus, one of the main mood-regulating regions of the brain. Moreover we report that these changes occurred in a context in which hippocampal TNF signaling was unbalanced toward TNFR1 due to increased expression of TNF and decreased levels of TNFR2. Finally, we showed that TNFR1−/− mice did not

Conclusions

Altogether, our data propose a novel molecular mechanism that regulates the development of depressive behavior associated with impaired hippocampal neurogenesis, neuronal and myelin plasticity and, that relies on hippocampal TNF signaling through TNFR1. We propose that therapeutic interventions based on the use of TNFR1-selective inhibitors may be beneficial not only to the treatment of neuropathic pain but also to the prevention of comorbid diseases such as depression and that this could occur

Conflict of interest

All authors declare that there are no conflicts of interest.

Acknowledgments

This work was supported by grants from the Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, from Ministero della Istruzione Università Ricerca (MIUR), under the Progetti di Interesse Nazionale (PRIN 2007) framework and from Fondazione Cariplo to Mariagrazia Grilli.

References (123)

  • A. Del Rey et al.

    Chronic neuropathic pain-like behavior correlates with IL-1beta expression and disrupts cytokine interactions in the hippocampus

    Pain

    (2011)
  • C. Ducottet et al.

    Effects of the selective nonpeptide corticotropin-releasing factor receptor 1 antagonist antalarmin in the chronic mild stress model of depression in mice

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2003)
  • V. Duric et al.

    Persistent pain produces stress-like alterations in hippocampal neurogenesis and gene expression

    J. Pain

    (2006)
  • H. Eyre et al.

    Neuroplastic changes in depression: a role for the immune system

    Psychoneuroendocrinology

    (2012)
  • E.S. Fu et al.

    Transgenic inhibition of glial NF-kappa B reduces pain behavior and inflammation after peripheral nerve injury

    Pain

    (2010)
  • A. George et al.

    Wallerian degeneration after crush injury of rat sciatic nerve increases endo- and epineurial tumor necrosis factor-alpha protein

    Neurosci. Lett.

    (2004)
  • J.G. Gleeson et al.

    Doublecortin is a microtubule-associated protein and is expressed widely by migrating neurons

    Neuron

    (1999)
  • L. Gonçalves et al.

    Neuropathic pain is associated with depressive behaviour and induces neuroplasticity in the amygdala of the rat

    Exp. Neurol.

    (2008)
  • N.A. Harrison et al.

    Neural origins of human sickness in interoceptive responses to inflammation

    Biol. Psychiatry

    (2009)
  • B.L. Hart

    Biological basis of the behavior of sick animals

    Neurosci. Biobehav. Rev.

    (1988)
  • P. Honore et al.

    Interleukin-1alphabeta gene-deficient mice show reduced nociceptive sensitivity in models of inflammatory and neuropathic pain but not post-operative pain

    Behav. Brain Res.

    (2006)
  • M.P. Kaster et al.

    Depressive-like behavior induced by tumor necrosis factor-alpha in mice

    Neuropharmacology

    (2012)
  • N. Kee et al.

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

    J. Neurosci. Methods

    (2002)
  • G. Kempermann et al.

    Genetic influence on phenotypic differentiation in adult hippocampal neurogenesis

    Brain Res. Dev. Brain Res.

    (2002)
  • G. Kempermann et al.

    Milestones of neuronal development in the adult hippocampus

    Trends Neurosci.

    (2004)
  • S. Kent et al.

    Sickness behavior as a new target for drug development

    Trends Pharmacol. Sci.

    (1992)
  • D. Kodama et al.

    Altered hippocampal long-term potentiation after peripheral nerve injury in mice

    Eur. J. Pharmacol.

    (2007)
  • R. Kolesnick et al.

    The sphingomyelin pathway in tumor necrosis factor and interleukin-1 signaling

    Cell

    (1994)
  • V.K. Kontinen et al.

    Behavioural measures of depression and anxiety in rats with spinal nerve ligation-induced neuropathy

    Pain

    (1999)
  • M. Kubera et al.

    Effects of serotonin and serotonergic agonists and antagonists on the production of tumor necrosis factor alpha and interleukin-6

    Psychiatry Res.

    (2005)
  • T. Lindenlaub et al.

    Effects of neutralizing antibodies to TNF-alpha on pain-related behavior and nerve regeneration in mice with chronic constriction injury

    Brain Res.

    (2000)
  • R.T. Martuscello et al.

    Increasing TNF levels solely in the rat hippocampus produces persistent pain-like symptoms

    Pain

    (2012)
  • L.A. McWilliams et al.

    Mood and anxiety disorders associated with chronic pain: an examination in a nationally representative sample

    Pain

    (2003)
  • J.M. Mettenburg et al.

    Diminished performance on neuropsychological testing in late life depression is correlated with microstructural white matter abnormalities

    Neuroimage

    (2012)
  • O. Mikova et al.

    Increased serum tumor necrosis factor alpha concentrations in major depression and multiple sclerosis

    Eur. Neuropsychopharmacol.

    (2001)
  • A.H. Miller et al.

    Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression

    Biol. Psychiatry

    (2009)
  • W. Pan et al.

    Increase in TNFα transport after SCI is specific for time, region, and type of lesion

    Exp. Neurol.

    (2001)
  • W. Pan et al.

    TNFα transport across the blood-brain barrier is abolished in receptor knockout mice

    Exp. Neurol.

    (2002)
  • W. Pan et al.

    Stroke upregulates TNFα transport across the blood-brain barrier

    Exp. Neurol.

    (2006)
  • K. Pfeffer et al.

    Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L monocytogenes infection

    Cell

    (1993)
  • K. Ren et al.

    Role of interleukin-1beta during pain and inflammation

    Brain Res. Rev.

    (2009)
  • R.M. Sapolsky

    Is impaired neurogenesis relevant to the affective symptoms of depression?

    Biol. Psychiatry

    (2004)
  • A.M. Aloisi et al.

    Effects of formalin-induced pain on ACTH, beta-endorphin, corticosterone and interleukin-6 plasma levels in rats

    Neuroendocrinology

    (1995)
  • H.A. Arnett et al.

    TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination

    Nat. Neurosci.

    (2001)
  • G. Bartzokis

    Neuroglialpharmacology: white matter pathophysiologies and psychiatric treatments

    Front. Biosci. (Landmark Ed)

    (2011)
  • P.A. Arnett et al.

    Depression in multiple sclerosis: review and theoretical proposal

    J. Int. Neuropsychol. Soc.

    (2008)
  • D. Balschun et al.

    Interleukin-6: a cytokine to forget

    FASEB J.

    (2004)
  • T.A. Bedrosian et al.

    Chronic dim light at night provokes reversible depression-like phenotype: possible role for TNF

    Mol. Psychiatry

    (2012)
  • J.M. Bessa et al.

    The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling

    Mol. Psychiatry

    (2009)
  • M. Bianchi et al.

    Interleukin-1 and nociception in the rat

    J. Neurosci. Res.

    (1998)
  • Cited by (0)

    1

    Address: Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, United States.

    View full text