Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation?

Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation?

Accepted Manuscript Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation? Nathan T. Fior...

1MB Sizes 43 Downloads 79 Views

Accepted Manuscript Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation? Nathan T. Fiore, Paul J. Austin PII: DOI: Reference:

S0889-1591(16)30103-9 http://dx.doi.org/10.1016/j.bbi.2016.04.012 YBRBI 2859

To appear in:

Brain, Behavior, and Immunity

Received Date: Revised Date: Accepted Date:

20 January 2016 11 April 2016 22 April 2016

Please cite this article as: Fiore, N.T., Austin, P.J., Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation?, Brain, Behavior, and Immunity (2016), doi: http://dx.doi.org/ 10.1016/j.bbi.2016.04.012

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Review article: Are the emergence of affective disturbances in neuropathic pain states contingent on supraspinal neuroinflammation?

Nathan T. Fiore, Paul J. Austin

Discipline of Anatomy & Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia

Corresponding author: Paul J. Austin

Laboratory of Neuroimmunology and Behaviour, Room E513, Anderson Stuart Building (F13), University of Sydney, NSW, 2006, Australia Email: [email protected] Telephone: +61 (0) 293515061

1

Abstract Neuro-immune interactions contribute to the pathogenesis of neuropathic pain due to peripheral nerve injury. A large body of preclinical evidence supports the idea that the immune system acts to modulate the sensory symptoms of neuropathy at both peripheral and central nervous system sites. The potential involvement of neuroimmune interactions in the highly debilitating affective disturbances of neuropathic pain, such as depression, anhedonia, impaired cognition and reduced motivation has received little attention. This is surprising given the widely accepted view that sickness behaviour, depression, cognitive impairment and other neuropsychiatric conditions can arise from inflammatory mechanisms. Moreover, there is also a set of well-described immune-to-brain transmission mechanisms that explain how peripheral inflammation can lead to supraspinal neuroinflammation. In the last 5 years increasing evidence has emerged that peripheral nerve injury induces supraspinal changes in cytokine or chemokine expression and alters glial cell activity. In this systematic review, based on strong preclinical evidence, we advance the argument that the emergence of affective disturbances in neuropathic pain states are contingent on pro-inflammatory mediators in the interconnected hippocampal-medial prefrontal circuitry that subserve affective behaviours. We explore how dysregulation of inflammatory mediators in these networks may result in affective disturbances through a wide variety of neuromodulatory mechanisms. There are also promising early findings that anti-inflammatory agents have efficacy to treat a variety of neuropsychiatric conditions including depression and appear suited to sub-groups of patients with elevated pro-inflammatory profiles. Thus, although further research is required, aggressively targeting supraspinal pro-inflammatory mediators at critical

2

time-points in appropriate clinical populations is likely to be a novel avenue to treat debilitating affective disturbances in neuropathic conditions.

Keywords Neuropathic pain, cytokines, astrocytes, microglia, neuroinflammation, hippocampus, medial prefrontal cortex, affective behaviour

3

1. Introduction Neuropathic pain is defined as ‘pain arising as a direct consequence of a lesion or disease affecting the somatosensory system’ (Treede et al., 2008), most commonly due to traumatic or iatrogenic injury, diabetes or infection. Clinically, neuropathic pain is characterised by both sensory and affective disturbances. Sensory disturbances include; spontaneous pain, allodynia, hyperalgesia, dysaesthesia and paraesthesia. The highly debilitating affective disturbances described by patients include impaired cognition, working memory dysfunction, decreased appetite, anhedonia, depression, disruption to sleep-wake cycles, as well as disturbances in familial and social interactions (Apkarian et al., 2004; Backonja et al., 2008; Bertolucci and de Oliveira, 2013; Dick and Rashiq, 2007; Fishbain et al., 1997; Hart et al., 2000; Jensen and Finnerup, 2007; Menefee et al., 2000; Meyer-Rosberg et al., 2001; Moriarty et al., 2011; Samwel et al., 2006). It should also be noted that the degree of disability associated with affective disturbances varies greatly between individuals, even in the case of post-surgical neuropathic pain cohorts where causality is less varied (Kehlet et al., 2006).

In the last 25 years researchers have developed four widely used preclinical nerve injury models in order to replicate the symptoms of neuropathic pain. They involve damaging a portion of axons which contribute to the sciatic nerve, and from greatest to least neuronal damage include: spinal nerve ligation (SNL), where the L5 and/or L6 spinal nerves are ligated (Kim and Chung, 1992); spared nerve injury (SNI), where the tibial and common peroneal sciatic nerve branches are tightly ligated then transected (Decosterd and Woolf, 2000); partial sciatic nerve ligation (PSNL), where approximately half of the sciatic nerve is ligated (Seltzer et al., 1990); and chronic

4

constriction injury (CCI), where four ligatures are loosely tied around the sciatic nerve (Bennett and Xie, 1988). In general these peripheral nerve injury models have similar time courses of sensory symptoms (emerging within 24 hours and persisting >2months), though signs of allodynia are greatest following SNL and smallest after CCI, and the most prominent behavioural signs of ongoing pain occur after CCI (for review see Kim et al., 1997). Further, the SNI model uniquely fails to produce thermal hyperalgesia, this may be explained by a lack of denervated Schwann cells which are known to produce many neuroactive molecules capable of acting on intact axons (for review see Costigan et al., 2009b; Decosterd and Woolf, 2000). Expression of affective disturbances, including anxiety- and depressive-like behaviours, sleep disturbances, anhedonia and changes in social interactions have been observed following all four nerve injury models, emerging approximately 1-2 weeks after injury, although there are also a number of studies which reported no changes in these behaviours (for review see Liu and Chen, 2014).

Nerve injury models, in combination with evoked-reflexive responses, have established that sensory disturbances, such as allodynia and hyperalgesia are a consequence of interactions between neurons, inflammatory immune and immunelike glial cells, as well as various immune-cell derived inflammatory mediators, in particular cytokines and chemokines, but also ATP, histamine, bradykinin and prostaglandins (PGs) (for review see Austin and Moalem-Taylor, 2010, 2013; Grace et al., 2014; Martini and Willison, 2016). Thus establishing neuropathic pain as a neuro-immune disorder. Nerve injury induces the release of inflammatory mediators at multiple levels of the neuraxis; strongest at the site of injury where it facilitates Wallerian degeneration and recovery, but also in the dorsal root ganglia (DRG) and

5

spinal cord recipient segments, and it is these peripheral and spinal sites of inflammation that appear to contribute to the development and maintenance of allodynia and hyperalgesia.

Peripheral neuropathy also induces production of inflammatory mediators within the brain, and this review will focus predominantly on preclinical findings of nerve injury induced alterations in expression of pro-inflammatory cytokines and glial cell activation markers. Although other inflammatory and chemical mediators have been implicated in pain neurobiology, changes in their expression patterns in the neuropathic brain have yet to be described. We will only briefly mention human findings mainly because brain-imaging technology is only just making it possible to investigate glial activation (Alshelh et al., 2016; Loggia et al., 2015), and analysis of cytokine expression profiles has only been examined through skin biopsies, or the collection of blood and CSF from chronic pain patients.

Surprisingly little attention has been paid to the potential role of supraspinal neuroinflammation in the emergence of affective disturbances in neuropathic pain states, which can be hypothesised on the basis of our understanding of the immune mechanisms of sickness behaviour, as well as evidence that cognitive impairment, depression and other neuropsychiatric disorders are associated with pro-inflammatory cytokines and chemokines (Capuron et al., 2002; Capuron and Miller, 2011; Capuron et al., 2005; Dantzer, 2009; Dantzer et al., 2008; Dowlati et al., 2010; McAfoose and Baune, 2009; Stuart and Baune, 2014).

6

In this review, we advance the argument that the emergence of affective disturbances in neuropathic pain states are contingent on altered inflammatory mediators in the interconnected hippocampal-medial prefrontal circuitry that subserves cognitive, emotional and motivational behaviours (i.e. affective behaviours). In our critical evaluation of the literature we find growing evidence that nerve injury evokes anatomically specific neuroinflammation in affective forebrain regions that is causally responsible for affective disturbances (Burke et al., 2013a; Dellarole et al., 2014; Mor et al., 2010; Narita et al., 2006; Nascimento et al., 2015; Norman et al., 2010b; Ren et al., 2011; Taylor et al., 2015; Wang et al., 2013a; Wu et al., 2014). Given the neural circuits mediating motivational, emotional and cognitive behaviours are highly conserved across species, we argue observations in these preclinical models are likely translatable to the human neuropathic pain experience (Craig, 2003; Keay et al., 2004; Murray et al., 2011). Thus the aim of this review is to systematically present the preclinical evidence that nerve-injury evoked supraspinal neuroinflammation underlies the development of affective disturbances by disrupting critical physiological processes. Following presentation of the preclinical evidence, we will discuss the clinical potential of anti-inflammatory treatments for neuropathic pain and other neuropsychiatric conditions.

2. Peripheral inflammation and affective disturbances It is well established that peripheral inflammation or an immune challenge in animals and humans triggers a set affective disturbances termed sickness behaviour (for review see Dantzer et al., 2008). Moreover, peripheral immune challenge appears to underlie modified behaviours through an anatomically specific pattern of supraspinal neuroinflammation, given both are antagonised by intracerebroventricular (i.c.v.)

7

injection of the anti-inflammatory cytokine interleukin-10 (IL-10) or minocycline treatment (Ban et al., 1992; Biesmans et al., 2013; Bluthe et al., 1999; Castanon et al., 2004; Gabellec et al., 1995; Henry et al., 2008; Laye et al., 2000; Laye et al., 1994; Siren et al., 2001; Utsuyama and Hirokawa, 2002).

Clinically, major depression disorder has a strong inflammatory component, with elevated blood levels of the pro-inflammatory cytokines tumor necrosis factor (TNF) and interleukin-6 (IL-6) in depressed individuals (Dowlati et al., 2010; Liu et al., 2012), and interferon-α (IFNα) immunotherapy is known to cause widespread neuropsychiatric symptoms (Capuron et al., 2002; Capuron et al., 2007; Pavol et al., 1995). Further, patients with painful neuropathy coupled with depression have been observed to have higher TNF levels than painful neuropathy patients without depression (Uceyler et al., 2007), an observation which supports the idea that comorbid symptoms of pain and depression may share a common inflammatory mechanism (Walker et al., 2014). Taking this idea a step further, we will now present evidence that an individual’s inflammatory response to injury or disease may predispose them to persistent pain and affective disturbances.

Keay and colleagues have developed a preclinical model to explore individual differences in neuropathic pain and disability using CCI combined with residentintruder social interactions testing and sleep-wake analysis in rats (Keay et al., 2004; Monassi et al., 2003). The major finding from these studies was that CCI leads to a persistent reduction in dominance behaviour and disrupted sleep-wake cycles in only a sub-group of rats, despite all nerve-injured rats having equivalent levels of allodynia. In a recent study utilising this model, we found that the sub-group of rats

8

with persistent disability in social interactions had significantly increased expression of T lymphocytes in the injured sciatic nerve compared to rats that displayed no affective disturbances (Austin et al., 2015). Within this disabled sub-group: IL-6 and the chemokine monocyte chemoattractant protein-1 (MCP-1, also known as CCL2) in the injured sciatic nerve, IL-6 in the DRG, and interleukin-1β (IL-1β) in the spinal cord were all selectively increased (Austin et al., 2015). Thus a distinct cytokine signature related to the enhanced adaptive immune response may underlie affective disturbances in this sub-group of nerve-injured rats.

3. Immune-to-brain signalling and inflammatory cytokines as neuromodulators Release of inflammatory cytokines may occur in response to the local environment in the central nervous system, but also due to the release of peripheral cytokines triggered by a distant inflammatory event (Mitchell et al., 2009). In the context of peripheral nerve injury, induction of MCP-1 and IL-1β compromise the integrity of the blood-spinal cord barrier increasing its permeability (Echeverry et al., 2011), which allows spinal cord infiltration of circulating T cells and monocytes (Cao and DeLeo, 2008; Costigan et al., 2009a; Fleming et al., 2009). In a more general context, the current understanding of immune-to-brain signalling is that three non-exclusive pathways exist; a) neural transmission, b) humoral transmission and c) molecular transmission (see Figure 1). We direct readers to the excellent recent review by Banks (2015) on the transmission of inflammatory mediators across the blood-brain barrier. When attempting to integrate the neural transmission route into the neuropathic setting, it should be noted that this could apply to an injured peripheral nerve, such as the sciatic nerve. Transmission would be via primary afferent fibres projecting to

9

dorsal horn neurons of the ascending anterolateral pathways, which in turn project to numerous supraspinal regions.

An emerging immune-to-brain transmission route could involve communication between the recently described ‘glymphatic system’ which enables passive circulation of interstitial fluid between the brain parenchyma and the subarachnoid CSF along paravascular spaces (Iliff et al., 2012; Yang et al., 2013), and the meningeal lymphatic vessels that carry leukocytes and drain into both deep and superficial cervical lymph nodes (Louveau et al., 2015).

Supraspinal cytokines and chemokines act in a neuromodulatory fashion, sharing similarities with neuropeptides, and operate under normal physiological conditions within a ‘cytokine network’ of neurons, microglia and astrocytes that are able to regulate cytokine production, express cytokine receptors and both amplify and attenuate cytokine signals (Haas and Schauenstein, 1997; Rothwell et al., 1996; Vitkovic et al., 2000). The magnitude of nerve-injury evoked supraspinal neuroinflammation is relatively mild compared to the peripheral site of nerve injury, or classical ‘neuroinflammation’ associated with an ischemic event, cell death or neurodegenerative disorders. Nevertheless, it appears to cause a perturbation in physiological cytokine concentrations sufficient to disrupt normal neuronal functioning, in what has been described as a ‘sub-inflammatory’ response (Adler et al., 2005; Adler and Rogers, 2005; Hutchinson and Watkins, 2014). A non-exhaustive summary of possible neuromodulatory mechanisms by which a dysregulation of inflammatory cytokines may underlie the development of affective behavioural changes is outlined in Figure 2. It is important to appreciate that the action of

10

inflammatory cytokines throughout the brain occurs in an anatomically specific fashion as cytokine and chemokine receptor expression is not ubiquitous, with regions critical for affective state regulation showing particularly strong expression patterns (i.e., hippocampus, hypothalamus, amygdala, nucleus accumbens and cortical regions) (Ban et al., 1991; Banisadr et al., 2002a; Banisadr et al., 2002b; Farrar et al., 1987; Gadient and Otten, 1993, 1994; Horuk et al., 1997; Kinouchi et al., 1991; Schöbitz et al., 1993; Takao et al., 1990; van der Meer et al., 2000; Yan et al., 1992).

4. Inflammatory cytokines and affective disturbances in the neuropathic brain Distinct anatomical regions of the hippocampus and mPFC, as well as interconnected regions such as the striatum/NAcc, ventral tegmental area (VTA), hypothalamus, amygdala and periaqueductal gray (PAG) are critical in regulating affective behaviours. Peripheral neuropathy produces elevated expression of pro-inflammatory cytokines and increased glial activation within these brain structures (Chu et al., 2012; Dellarole et al., 2014; Marcello et al., 2013; Mor et al., 2010; Narita et al., 2006; Norman et al., 2010b; Ren et al., 2011; Takeda et al., 2009; Taylor et al., 2015; Wang et al., 2013a; Wu et al., 2014) (see Table 1). In addition there is emerging evidence that neuroinflammation in these forebrain and midbrain regions leads to the expression of affective disturbances (Burke et al., 2013a; Dellarole et al., 2014; Mor et al., 2010; Narita et al., 2006; Nascimento et al., 2015; Norman et al., 2010b; Ren et al., 2011; Taylor et al., 2015; Wang et al., 2013a; Wu et al., 2014) (see Table 2). It should be noted that in the mPFC and the hippocampus, as well as in the ventral posterolateral thalamus and brainstem structures (i.e., the locus coeruleus, rostroventral medulla, dorsal column nuclei and red nucleus), nerve injury-induced neuroinflammation has been causally linked with allodynia and hyperalgesia (Chiang

11

et al., 2013; Covey et al., 2000; del Rey et al., 2011; Gosselin et al., 2010; Guo et al., 2012; Huang et al., 2014; Ignatowski et al., 1999; Ignatowski et al., 2005; LeBlanc et al., 2011; Li et al., 2008; Liu et al., 1995; Spengler et al., 2007; Tsai et al., 2012; Wang et al., 2015a; Wang et al., 2008; Wang et al., 2012; Wei et al., 2008; Zeng et al., 2014). With that said, the role of supraspinal neuroinflammation in the development of sensory dysfunction after nerve injury is not the focus of this review, and thus will not be discussed in detail. We hypothesise that peripheral nerve injury produces a specific pattern of supraspinal neuroinflammation that provides a basis for modulation of the neuroanatomical circuits controlling affective behaviours, thus contributing to the affective disturbances associated with neuropathic pain states. We will now examine the hippocampus, mPFC, striatum/NAcc, VTA, hypothalamus, amygdala and PAG in turn, briefly considering the role of each in affective disturbances after nerve injury before discussing how neuroinflammation can influence these affective disturbances.

4.1 Hippocampus The hippocampus modulates cognition and emotional state with dorsal and ventral functional networks influencing these two aspects of behaviour respectively (for detailed review see Fanselow and Dong, 2010; Moser and Moser, 1998). Hippocampal neurons are activated by noxious stimuli and their role in pain perception and awareness is longstanding (Dutar et al., 1985; Khanna and Sinclair, 1989). Recent preclinical studies have identified structural and functional changes in the hippocampus following experimental nerve injury (Cardoso-Cruz et al., 2011; Dellarole et al., 2014; Dimitrov et al., 2014; Hu et al., 2010; Kalman and Keay, 2014; Kodama et al., 2007; Mutso et al., 2012; Tanabe et al., 2008; Taylor et al., 2014;

12

Terada et al., 2008). These nerve injury evoked changes in hippocampal anatomy and function appear to underpin disturbances in hippocampal-dependent cognition (Cardoso-Cruz et al., 2013; Dimitrov et al., 2014; Gregoire et al., 2012; Hu et al., 2010; Kodama et al., 2011; Leite-Almeida et al., 2012; Mutso et al., 2012; Ren et al., 2011; Wang et al., 2013a) and emotion (Dimitrov et al., 2014; Fukuhara et al., 2012; Gregoire et al., 2012; Hu et al., 2010; Leite-Almeida et al., 2012; Mutso et al., 2012; Nascimento et al., 2015; Wang et al., 2015b).

Over the last 15 years it has emerged that there is a robust upregulation of proinflammatory cytokines, TNF, IL-1β and IL-6 in the hippocampi of neuropathic rodents, strongest at 1-2 weeks and persisting beyond 3 weeks after injury (Al-Amin et al., 2011; Covey et al., 2000; del Rey et al., 2011; Gerard et al., 2015; Ignatowski et al., 1999; Ren et al., 2011; Sud et al., 2008; Uceyler et al., 2008). The cellular origins of hippocampal cytokines have yet to be fully evaluated, however nerve injury is known to increase the expression of TNF in hippocampal neurons (Spengler et al., 2007; Wang et al., 2013a). A glial origin is also possible although evidence is limited to only a single study showing SNL increased the expression of the astrocyte activation marker GFAP but only when combined with early life stress in male rats, whilst common peroneal nerve ligation did not alter microglia density or phenotype (Burke et al., 2013b; Zhang et al., 2008).

Regardless of the cellular origin, there is growing evidence that TNF, IL-1β and IL-6 are involved in altered hippocampal function and consequent affective disturbances after nerve injury. TNF in particular has emerged as a key player in injury-evoked impairment of LTP, reduced neurogenesis and decreased plasticity, evidenced through

13

reduced expression of synaptic and myelin proteins (Dellarole et al., 2014; Gerard et al., 2015; Ren et al., 2011; Spengler et al., 2007; Sud et al., 2008; Wang et al., 2013a). Maternal deprivation (MD) combined with nerve injury exacerbated allodynia and increased expression of IL-6 and TNF in females rats, whilst in male rats there was an increase in GFAP and IL-1β as well as an increase in anxiety-like behaviour (Burke et al., 2013b). Similarly, early-life stress has been demonstrated as a risk factor for chronic pain clinically (Davis et al., 2005).

Hippocampal TNF and IL-1β are known to modulate glutamatergic signalling adversely affecting cognition. TNF alone enhances the expression of AMPA receptors lacking GluR2 expression leading to enhanced calcium currents, whilst TNF reduces the currents induced by glutamate, NMDA, AMPA and kainate (Beattie et al., 2002; Furukawa and Mattson, 1998; Stellwagen et al., 2005; Wang et al., 2013a). IL-1β reduces spontaneous excitatory post-synaptic currents in hippocampal neurons, but enhances NMDA-mediated currents that promote increased neurotoxicity (Viviani et al., 2003; Yang et al., 2005). Hippocampal LTP via NMDA-receptors underpins new memory formation, however TNF and IL-1 have well-established inhibitory effects on LTP (Butler et al., 2004; Cunningham et al., 1996; Tancredi et al., 1992). Thus is it perhaps unsurprising that elevated TNF in the dorsal hippocampus of nerve-injured rats suppresses NMDA mediated-currents, reducing LTP and consequently leading to impaired short-term memory (Ren et al., 2011; Wang et al., 2013a).

Neuroinflammation also appears to modulate affective behaviours via NMDA receptors. TNF-induced development of depressive-like behaviour is reversed by agmatine, an endogenous amine that blocks NMDA receptors (Neis et al., 2014). In

14

neuropathic rats NMDA receptor activation with D-serine was anxiolytic, which may be explained by reduced expression of NR1 and NR2B subunits in the ventral hippocampus in the neuropathic setting (Wang et al., 2015b). Interestingly, D-serine had no effect on pain sensitivity, which is supportive of the idea that NMDAblockade not activation reduces pain (Sarkis et al., 2011; Wang et al., 2015b). NMDA receptor function is clearly implicated in mediating hippocampal-dependent cognitive function and affective behaviours, and nerve injury evoked TNF is a strong candidate to mediate NMDA receptor dysfunction.

Hippocampal neurogenesis is integral for affective state regulation (Sahay and Hen, 2007), however hippocampal TNF has been shown to inhibit this process (Cacci et al., 2005; Monje et al., 2003). In the context of nerve injury increased hippocampal TNF, acting on TNFR1, results in reduced neurogenesis and plasticity, as well as anhedonia and depressive-like behaviours (Dellarole et al., 2014; Nascimento et al., 2015). These behaviours were normalised by fluoxetine and thalidomide treatments that reduced TNF. Pro-inflammatory cytokines may contribute to a reduction in neurogenesis by suppression of the critical neurotrophic factor BDNF. Low levels of BDNF in the hippocampus are associated with stress, depression and pain (Duric and McCarson, 2005; Duric and McCarson, 2006; Duric and McCarson, 2007; Fukuhara et al., 2012). IL-1β reduces the production of BDNF, impairs neurogenesis and leads to sedentary behaviour and anhedonia (Goshen et al., 2008; Koo and Duman, 2008). In neuropathic rats elevated IL-6 expression mirrored a decrease in BDNF, both of which were reversed by blocking NMDA receptors (Al-Amin et al., 2011). Further neuropathy-induced impaired spatial memory, depressive-like behaviour and decreased BDNF expression were all normalised by amitriptyline, which is known to

15

reverse elevated hippocampal TNF (Hu et al., 2010; Sud et al., 2008). It should be noted however, elevated spinal BDNF is pronociceptive after nerve injury (Coull et al., 2005), thus BDNF may not be a suitable target to normalise neuropathic symptoms, owing to different functions across the neuraxis.

NF-κB is a transcription factor activated by TNF and other cytokines. It is heavily involved in the transcription of genes encoding pro-inflammatory cytokines, and after nerve injury its role in pain facilitation at the spinal cord level is well documented (Ledeboer et al., 2005). Nerve injury induces an increase in hippocampal expression of the NF-κB subunit p65 that is ameliorated by NMDA receptor blockade (Chou et al., 2011). Activation of NF-κB also reduces BDNF resulting in impaired neurogenesis and depressive-like behaviours (Goshen et al., 2008; Haydon and Carmignoto, 2006; Koo and Duman, 2008). Thus, NF-κB may be a critical molecule in modulating signalling through NMDA receptors and suppressing BDNF expression. In summary, neuroinflammation in the neuropathic hippocampus is likely to influence affective behaviours by modulation of NMDA receptor function and/or reduction of neurogenesis via suppression of BDNF, whilst inhibition of LTP, also via NMDA receptors, is the likely mechanism of impaired memory.

4.2 Prefrontal cortex The rat prefrontal cortex (PFC) is an area of association cortex that predominantly consists of the mPFC and the orbitofrontal cortex (OFC) and regulates cortical control of visceral functions, as well as taking part in high-level cognitive and emotional processing (Ongur and Price, 2000). In the PFC nerve injury has been shown to induce anatomical and epigenetic changes, as well as dysregulation of glutamatergic

16

signalling (Metz et al., 2009; Seminowicz et al., 2009; Tajerian et al., 2013). Of the mPFC sub-divisions three have been linked with affective disturbances in neuropathic conditions, they are from dorsal to ventral, the anterior cingulate (ACC), the prelimbic (PL) and infralimbic (IL) cortices. These nerve injury evoked changes appear to underpin disturbances in ACC-dependent cognition and motivation (Cardinal et al., 2003; LaBuda and Fuchs, 2005; LaGraize et al., 2004; Qu et al., 2011; Rajasethupathy et al., 2015; Walton et al., 2003) as well as PL/IL-dependent cognition and emotion (Giordano et al., 2012; Roozendaal et al., 2004). Currently, there is a lack of evidence that the OFC is involved in affective disturbances after nerve injury.

In analysing the PFC changes in neuroinflammation after nerve injury most of the early studies used a large tissue block (Apkarian et al., 2006; Liu et al., 2007; Uceyler et al., 2008), whilst more recent studies have focused on sub-divisions of the mPFC or the OFC (Al-Amin et al., 2011; Fuccio et al., 2009; Giordano et al., 2012; Shao et al., 2015). The consensus of these studies has been that nerve injury induces a proinflammatory response in the PFC that is most apparent 1-2 weeks post-injury and could be responsible for the neuroplastic changes described above. Moreover, there is growing evidence that PFC neuroinflammation is causally related to affective disturbances. Nerve injury induced elevation of IL-1β in the PFC results in depressive-like behaviour, which could be reversed by i.c.v. injection of either IL-1ra or oxytocin, with the latter attenuating IL-1β expression (Norman et al., 2010a; Norman et al., 2010b). Elevated TNF in the PFC also leads to depressive-like symptoms, both of which can be reversed by thalidomide or fluoxetine treatment (Nascimento et al., 2015). Chronic minocycline treatment led to a reduction in pro-

17

inflammatory M1 microglia in the PFC of nerve-injured rats, whilst promoting polarisation to the anti-inflammatory M2 microglia in nerve-injured and olfactory bulbectomised (OB) depressed-rats (Burke et al., 2014). Despite the two different effects on microglia in the presence or absence of a depressive phenotype, minocycline reduced sensory disturbances in both cases, as well as normalising depressive-like behaviour in OB-SNL rats (Burke et al., 2014).

The ACC has been the focus of many studies due to its importance in encoding the affective component of pain, and nerve injury has been shown to increase IL-1β and IL-6 expression and reduce BNDF expression in this region (Al-Amin et al., 2011). Although these changes have not been directly linked to disruption in affective behaviours they were prevented by NMDA receptor blockade. Given that activation of NMDA receptors in the ACC mediates conditioned-place aversion (CPA) and supresses BNDF synthesis (Johansen and Fields, 2004; Kim et al., 2009), it is likely expression of CPA, upregulation of inflammatory mediators and suppression of BDNF are all dependent on NMDA receptor activation in the ACC.

Glial activity in the ACC has been directly linked to glutamate signalling and modulation of affective behaviours, with ipsilateral ACC astrocyte activation accompanied by anxiety-like behaviour 4 weeks after PSNL (Narita et al., 2006). Further investigation revealed ACC changes in astrocyte morphology led to increased glutamate and decreased GABA, and that astrocyte activation was causally related to sleep disturbances post-injury (Narita et al., 2011; Yamashita et al., 2014). Moreover, nerve injury enhances escape/avoidance behaviour, which was significantly inhibited via activation of GABAA receptors in the ACC (LaGraize et al., 2004).

18

In the PL/IL, nerve injury increased IL-1β expression in astrocytes and IL-1R1 expression in glutamatergic neurons, alongside elevated extracellular glutamate, increased expression of vesicular and glial glutamate and GABA transporters (de Novellis et al., 2011; Giordano et al., 2012; Marcello et al., 2013). Altered fearresponding as a consequence of nerve injury-induced changes in glutamate neurotransmission in the PL/IL could indeed be driven by pro-inflammatory cytokines, since TNF, IL-1β and IFNγ have previously been shown to increase glutamate release from astrocytes (Ida et al., 2008).

Clearly there is a complex bi-directional modulation between pro-inflammatory cytokines and several neurotransmitter systems in the mPFC, however elevated glutamate possibly leading to cell death, decreased GABA and BDNF, and cytokine induction downstream of NMDA receptors are some common features. Further, there is increasing evidence that neuroinflammation in the neuropathic PFC leads to affective disturbances, including anxiety- and depressive-like behaviours as well as disruption to sleep/wake cycles.

4.3 Striatum and Nucleus accumbens The dorsal striatum comprises the caudate and putamen in humans, a single structure in the rat, and is primarily involved in action selection of cognitive, motor and oculomotor functions (Alexander et al., 1986), whereas the ventral striatum largely consists of the NAcc, which regulates motivational drive and goal-directed behaviour (Brischoux et al., 2009; Salamone et al., 2007; Sugam et al., 2012).

19

Nerve injury alters the functional connectivity of the NAcc (Baliki et al., 2014; Chang et al., 2014), as well as myriad changes within the dopaminergic system (Austin et al., 2010; Chang et al., 2014; Sagheddu et al., 2015; Taylor et al., 2015; Wu et al., 2014). Nerve injury also impairs motivation and emotion, which are associated with changes in the glutamatergic system of the NAcc (Goffer et al., 2013; Marcello et al., 2013; Schwartz et al., 2014).

Nerve injury has been reported to produce a pro-inflammatory cytokine response in both aspects of the striatum. TNF is increased bilaterally in the NAcc of rats a week after SNI (Wu et al., 2014). In the contralateral striatum/thalamus, IL-1β mRNA is decreased 10 days after CCI and SNI, however by day 24 there is an increase in the SNI group (Apkarian et al., 2006). Another study reported a bilateral increase in striatal IL-1β protein expression 2 weeks after both SNI and CCI, changes which are co-incident with a reduction in striatal BDNF (Al-Amin et al., 2011). Further blockade of accumbal NMDA receptors, or activation of dopamine receptors attenuated the increase in IL-1β, as well as normalising BDNF levels and allodynia (Al-Amin et al., 2011; Sarkis et al., 2011).

Although the NAcc is critical for regulating affective disturbances after nerve injury there is only one study that conclusively demonstrates the involvement of neuroinflammation. TNF was found to be upregulated in the NAcc after SNI, alongside increased DAT expression and decreased synaptic dopamine levels (Wu et al., 2014). Behaviourally this was associated with prevention of conditioned place preference (CPP) to low dose morphine, indicating a reduction in reward-seeking behaviour. Moreover, neutralising TNF or knocking out TNFR1 normalised both

20

DAT expression and synaptic dopamine levels, and reinstated CPP. Thus, post-injury accumbal release of TNF acting via TNFR1 appears to play a role in reduced motivation (Wu et al., 2014). Elevated striatal or accumbal cytokines may play additional roles in affective behaviours, given their ability to modulate both glutamatergic and dopaminergic signalling, both of which are disrupted and associated with affective disturbances after nerve injury in these brain regions.

4.4 Ventral tegmental area (VTA) The VTA is a crucial part of the brains reward circuitry (Lammel et al., 2011; Lammel et al., 2014; Lammel et al., 2012). Nerve injury impairs the opioid and dopaminergic systems of the VTA, which results in disturbances in VTA-dependent motivation (Ewan and Martin, 2011a, b; Martin et al., 2007; Ozaki et al., 2002; Taylor et al., 2015; Wu et al., 2014).

Based on the single study to investigate expression of pro-inflammatory cytokines in the VTA there was no change in TNF expression a week after nerve injury (Wu et al., 2014). Peripheral nerve injury, where the sciatic nerve is encased in polyethylene tubing, has however been shown to increase microglial activation in the VTA, leading to decreased opioid- and cocaine-stimulated dopamine responses resulting in blockade of CPP (Taylor et al., 2015). The authors suggest this occurs through loss of expression of the chloride transporter, KCC2, in GABAergic interneurons of the VTA, which leads to disinhibition of their projection targets. This study therefore highlights the potential of microglial inhibitors such as minocycline to normalise affective disturbances in the neuropathic state. Further studies are required to

21

elucidate the exact mechanism of microglial-neuronal interaction, as TNF does not appear to be involved (Wu et al., 2014).

4.5 Amygdala The amygdala comprises a critical part of the circuitry regulating negatively valenced emotion, and is also implicated in cognition and motivation (Amano et al., 2010; Cai et al., 2014; Fanselow and Poulos, 2005; Janak and Tye, 2015; Jennings et al., 2013; Kim et al., 2013; Knobloch et al., 2012; Orsini et al., 2015; Penzo et al., 2014; Petrovich et al., 2001; Stefanik and Kalivas, 2013). Nerve injury leads to anatomical changes within the amygdala (Goncalves et al., 2008), as well as changes in glutamatergic and GABAergic activity that is co-incident with alterations in motivation and emotion (Ansah et al., 2010; Pedersen et al., 2007).

Few studies have examined changes in neuroinflammation in the neuropathic amygdala and those that have yielded mixed results. Reactive astrogliosis, but no change in microglial activity, density or phenotype, has been reported a week after nerve injury (Marcello et al., 2013; Zhang et al., 2008). Astrogliosis does occur alongside increased glial glutamate transporter-1, glial glutamate aspartate reuptake transporter and vesicular glutamate transporter 1, at a time-point when anxiety-like behaviour is evident, however no causal relationship has been demonstrated (Burke et al., 2013a; Marcello et al., 2013). By three weeks after nerve injury there is a predominantly anti-inflammatory response, with an increase in IL-10 and a decrease in IL-6 gene expression, with no change in IL-1β, TNF, GFAP and IBA-1 (Burke et al., 2013a). Further studies are urgently needed to investigate whether amygdala astrogliosis and a delayed anti-inflammatory response are involved in affective

22

disturbances, possibility through dysregulation of glutamatergic and GABAergic systems also reported in the neuropathic amygdala.

4.6 Hypothalamus The hypothalamus plays an essential role in coordinating neuroendocrine, autonomic, and behavioural responses, as well as the sleep/wake cycle (for detailed review see Swanson, 2000). The hypothalamus and HPA axis are important in regulating affective behaviours after nerve injury (Norman et al., 2010b). Nerve injury has been reported to increase corticosterone levels (Kilburn-Watt et al., 2010), although contrastingly no changes in hypothalamic neuronal activation markers or CRH expression in basal or stress stimulatory conditions were reported (Bomholt et al., 2005; Ulrich-Lai et al., 2006). Nerve injury has been shown to lead to disturbances in the hypothalamic-pituitary-thyroid (HPT) axis in a sub-group of rats with persistent disability in social interactions after CCI (Kilburn-Watt et al., 2010, 2014).

Relatively few studies have examined expression of pro-inflammatory cytokines in this region in neuropathic rodents, yielding largely no change or a reduction in cytokine expression (Kilburn-Watt et al., 2014; Uceyler et al., 2008). A role for hypothalamic neuroinflammation in modulating nerve injury induced affective disturbances is limited, given the expression of disability in social interactions and dysregulation of the HPT axis were unrelated to changes in hypothalamic IL-1β and TNF measured 7 days after CCI (Kilburn-Watt et al., 2014). Although previous studies using peripheral immune activation demonstrated increased hypothalamic IL1β expression was capable of supressing the HPT axis at much earlier time points (Boelen et al., 2004; Boelen et al., 2006; Kakucska et al., 1994). Moreover, an

23

increase in hypothalamic microglial activation was reported 4 days after CCI, which was blocked by an NMDA receptor antagonist (Takeda et al., 2009). Thus it remains a possibility that expression of hypothalamic neuroinflammation in the first few days following nerve injury may lead to affective disturbances.

4.7 Periaqueductal gray The PAG mediates descending pain modulation and integrated emotional coping strategies, as well as sleep-wake cycles (Bandler and Shipley, 1994; Gerashchenko et al., 2003; Gerashchenko et al., 2001; Gerashchenko and Shiromani, 2004; Keay and Bandler, 2001; Lu et al., 2006). In the PAG nerve injury has been shown to alter glutamatergic signalling (Marcello et al., 2013; Terashima et al., 2012), an effect which appears to be specific to the ventrolateral PAG column (vlPAG) (Ho et al., 2013; Renno, 1998).

There are mixed findings regarding neuroinflammation in the PAG after nerve. Firstly, nerve injury in rats increases expression of TNF, IL-1β and IL-6 in the PAG, effects which were reversed by a selective inhibitor of melanocortin-4 receptor, which also increased IL-10 and normalised sensory disturbances, perhaps through altered descending pain modulation (Chu et al., 2012). Whereas in mice, there was no change in IL-1β, IL-6 or TNF expression after SNI (Norman et al., 2010a). PSNL and CCI lead to increased microglia expression 4 and 6 days after injury, whereas a week after SNI and common peroneal nerve ligation there was no change in microglia density or activation (Marcello et al., 2013; Takeda et al., 2009; Zhang et al., 2008). Increased expression of astrocyte activation markers has been demonstrated in the PAG a week after CCI, however there have been conflicting results following SNI (Chu et al.,

24

2012; Marcello et al., 2013; Mor et al., 2010; Norman et al., 2010b). Glial activation may underlie some changes in affective behaviours after SNI, as increased GFAP in the PAG occurred in mice with depressive-like behaviour (Norman et al., 2010b). Studies by Mor and colleagues have demonstrated columnar specificity of neuroinflammation with increased GFAP as well as evidence of cell death localised to the lateral PAG and caudal vlPAG columns of the PAG in a sub-group of rats that show persistent disability in social interactions and sleep disturbances after CCI (Monassi et al., 2003; Mor et al., 2011; Mor et al., 2010). This same sub-group of rats have decreased glucocorticoid receptor (GR) within the vlPAG column that is responsible for passive coping strategies and sleep regulation, but increased GR in the dorsolateral PAG, which mediates active coping strategies in response to psychological stressors (Mor and Keay, 2013). Therefore anatomically specific glial and neuronal maladaptations in the PAG after nerve injury appear to drive affective and sleep disturbances in this subgroup of rats. Thus, the best evidence for the involvement of neuroinflammation in the PAG with affective disturbances are the glial activation patterns specific to the vlPAG.

5. Anti-inflammatory treatments in neuropathic pain Anti-inflammatory treatments which interfere with pro-inflammatory cytokines or block glial activation have shown success in reducing allodynia and hyperalgesia in preclinical neuropathic pain models, and blockers of TNF, IL-1β and IL-6 are all effective in treating inflammatory pain conditions, such as rheumatoid arthritis (for detailed review see Austin and Moalem-Taylor, 2010; Austin and Moalem-Taylor, 2013; Kwilasz et al., 2015). Clinically, numerous studies have reported that patients with painful neuropathy have a lower expression of anti-inflammatory cytokines

25

within the CSF and blood (Backonja et al., 2008; Uceyler et al., 2007), which suggests that an insufficiency of anti-inflammatory cytokines may propagate ongoing inflammation as well as clinical symptoms associated with the neuropathic pain state. Therefore, increasing physiological levels of anti-inflammatory cytokines may be a successful strategy to reduce inflammation and the associated sensory and affective disturbances in the neuropathic pain state. Despite this several randomised controlled drug trials have reported a lack of efficacy of novel anti-inflammatories to treat neuropathic pain (Bramson et al., 2015; Kalliomaki et al., 2013; Knezevic et al., 2015; Landry et al., 2012). This poor translational outcome is likely due in part to the problematic nature of drug delivery, which usually has a transient efficacy and can display unwanted side effects associated with the higher doses needed to effectively block the pain response. Further, in preclinical models anti-inflammatories are administered in the acute phase shortly after nerve injury when the contribution of pro-inflammatory mediators to allodynia and hyperalgesia are greatest, whereas in patients with established chronic pain (>6 months) the contribution of peripheral inflammatory mediators is diminished. Recent developments in anti-inflammatory cytokine-based gene therapies provide a novel approach to increasing the physiological expression of anti-inflammatory cytokines for a prolonged period of time, and have been found to be highly efficacious in preclinical models of chronic pain (for detailed reviews on anti-inflammatory gene therapies in chronic pain see Goins et al., 2012; Kwilasz et al., 2015). Of particular note, encapsulating plasmid DNA encoding IL-10 in biodegradable polymer-based micro-particles has been successful in producing sustained therapeutic effects, requiring a fraction of the dose of plasmid DNA encoding IL-10 compared to when the plasmid DNA was injected naked (Sloane et al., 2009; Soderquist et al., 2010). XT-101 and XT-150,

26

encapsulated IL-10 gene therapy products developed by Xalud Therapeutics, appear to be the leading candidates for the delivery of IL-10 for prolonged periods and treatment for neuropathic pain, though results from clinical trials are not yet available.

Differences in the time-course and anatomical location of peripheral inflammation compared to supraspinal neuroinflammation in neuropathic pain states enhance the possibility that dampening the latter may successfully alleviate affective disturbances. The preclinical findings presented throughout this review confirm that assertion, however clinically the effectiveness of anti-inflammatory treatments to reduce affective disturbances associated with neuropathic pain have yet to be investigated. With that said randomised controlled trials have proven anti-inflammatories to be effective in treating depression. The cyclooxygenase-2 inhibitor, celecoxib, is effective in combination with antidepressants (Abbasi et al., 2012; Akhondzadeh et al., 2009; Majd et al., 2015; Muller et al., 2006), whilst the cytokine blocker, infliximab a soluble TNF antibody, is effective in treating major depressive disorder in a sub-group of patients with elevated C-reactive protein (Raison et al., 2013). Minocycline, which blocks microglial activation, has shown some early promise in the treatment of several neuropsychiatric disorders when combined with antidepressants or antipsychotics, although randomised controlled trials have yet to be completed (Dean et al., 2012; Miyaoka et al., 2012; Miyaoka et al., 2008).

Interestingly, there is preclinical evidence that dampening neuroinflammation in affective brain regions leads to a reduction in sensory disturbances, although affective disturbances were not tested in these studies (Covey et al., 2000; del Rey et al., 2011; Ignatowski et al., 1999; Ignatowski et al., 2005; Spengler et al., 2007). These findings

27

indicate either a dual role for these regions in regulating both affective and sensory aspects of pain, or that reversal of affective disturbances improves the perception of sensory disturbances. Future studies whether clinical or preclinical should therefore examine both affective and sensory disturbances when exploring the role of neuroinflammation in neuropathic pain to delineate the specific mechanisms of each. Although further research is required the exciting possibility remains that aggressively targeting supraspinal inflammatory mediators in appropriate clinical populations, either alone, or in combination with current first-line treatments, may be efficacious in treating debilitating affective disturbances associated with neuropathic pain states.

6. Conclusions Following our systematic evaluation of the preclinical evidence, we assert that the emergence of affective disturbances in neuropathic pain states are contingent on altered inflammatory mediators in the interconnected hippocampal-medial prefrontal networks that subserve affective behaviours. Further our recent preclinical findings suggest that an individual’s immune reactivity could predispose them to pain and affective disturbances. Therefore, certain clinical populations may be more responsive to potential anti-inflammatory treatments.

After nerve injury transmission of peripheral inflammation to the brain through a set of well-defined immune-to-brain signalling pathways likely leads to supraspinal neuroinflammation. Nerve injury evokes an anatomically specific pattern of neuroinflammation. We believe neuroinflammation in the interconnected hippocampal-mPFC circuitry is critical in the development of depression, anhedonia,

28

cognitive impairment and sleep disturbances that are a common feature of neuropathic pain in both preclinical and clinical settings. It is also possible that once neuroinflammation is established in these heavily interconnected circuits it may be capable of self-amplification by volume diffusion or neural transmission that may underlie the entrenchment of affective disturbances.

There are a variety of compelling mechanisms by which nerve injury induced neuroinflammation modulates neural circuits controlling affective behaviours. The strongest evidence is for modulation of the glutamatergic system, either directly via elevated glutamate levels, altered expression of NMDA and AMPA receptors and glutamate transporters, or indirectly through the metabolites of the IDO pathway. Major consequences of this are disruption of LTP, which has been linked to cognitive deficits, whilst metabolites of the IDO pathway and elevated glutamate have been linked with neurotoxicity and depression. Suppression of critical brain neurotrophic factors, such as BDNF, by cytokines may contribute to a reduction in neurogenesis and neuroplasticity, processes which are diminished in depression. TNF modulation of dopaminergic systems has been associated with blunted reward, whilst midbrain glial activation appears to contribute to midbrain cell death and altered expression of glucocorticoids that is associated with affective disturbances.

Despite mounting evidence of a causal relationship between neuroinflammation in affective brain regions after peripheral nerve injury and affective disturbances preclinically, a greater appreciation of several key areas is still required. In order to fully support the notion that nerve injury-evoked inflammatory mediators in brain regions associated with affective behaviours underpin affective disturbances, a more

29

detailed anatomical examination of their expression patterns and cellular origin is necessary. Further, the exact timeline for the appearance of supraspinal neuroinflammation in different regions remains to be elucidated. To address this, longitudinal studies examining the magnitude of neuroinflammation throughout the brain in several nerve injury models combined with thorough assessment of sensory and affective disturbances are required. These preclinical studies should be combined with longitudinal brain imaging studies in patients with neuropathic pain that take advantage of new imaging techniques that can examine glial activation patterns(Alshelh et al., 2016; Loggia et al., 2015). Developing a greater appreciation of the precise molecular mechanisms and anatomical circuits through which neuroinflammation regulates specific facets of affective behaviour in neuropathic pain states should also be a priority. Understanding these key issues is critical to guide the development of novel strategies to target specific supraspinal inflammatory mediators at critical time-points in appropriate clinical populations, thus enhancing the potential for efficacious treatments of the usually treatment-resistant affective disturbances.

Acknowledgements The authors would like to thank Dr. F.R. Walker for his critical appraisal and insight on an earlier version of the manuscript.

30

References Abbasi, S.-H., Hosseini, F., Modabbernia, A., Ashrafi, M., Akhondzadeh, S., 2012. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: Randomized double-blind placebo-controlled study. Journal of affective disorders 141, 308-314. Adler, M.W., Geller, E.B., Chen, X., Rogers, T.J., 2005. Viewing chemokines as a third major system of communication in the brain. The AAPS journal 7, E865-870. Adler, M.W., Rogers, T.J., 2005. Are chemokines the third major system in the brain? Journal of leukocyte biology 78, 1204-1209. Akhondzadeh, S., Jafari, S., Raisi, F., Nasehi, A.A., Ghoreishi, A., Salehi, B., Mohebbi-Rasa, S., Raznahan, M., Kamalipour, A., 2009. Clinical trial of adjunctive celecoxib treatment in patients with major depression: a double blind and placebo controlled trial. Depression and Anxiety 26, 607-611. Al-Amin, H., Sarkis, R., Atweh, S., Jabbur, S., Saade, N., 2011. Chronic dizocilpine or apomorphine and development of neuropathy in two animal models II: Effects on brain cytokines and neurotrophins. Experimental neurology 228, 30-40. Alexander, G.E., DeLong, M.R., Strick, P.L., 1986. Parallel organization of functionally segregated circuits linking basal ganglia and cortex. Annual review of neuroscience 9, 357-381. Alshelh, Z., Di Pietro, F., Youssef, A.M., Reeves, J.M., Macey, P.M., Vickers, E.R., Peck, C.C., Murray, G.M., Henderson, L.A., 2016. Chronic Neuropathic Pain: It's about the Rhythm. The Journal of Neuroscience 36, 1008-1018. Amano, T., Unal, C.T., Pare, D., 2010. Synaptic correlates of fear extinction in the amygdala. Nature neuroscience 13, 489-494. Ansah, O.B., Bourbia, N., Goncalves, L., Almeida, A., Pertovaara, A., 2010. Influence of amygdaloid glutamatergic receptors on sensory and emotional painrelated behavior in the neuropathic rat. Behavioural brain research 209, 174-178. Apkarian, A.V., Lavarello, S., Randolf, A., Berra, H.H., Chialvo, D.R., Besedovsky, H.O., del Rey, A., 2006. Expression of IL-1beta in supraspinal brain regions in rats with neuropathic pain. Neuroscience letters 407, 176-181. Apkarian, A.V., Sosa, Y., Krauss, B.R., Thomas, P.S., Fredrickson, B.E., Levy, R.E., Harden, R.N., Chiavlo, D.R., 2004. Chronic pain patients are impaired on an emotional decision-making task. Pain 108, 129-136. Austin, P.J., Berglund, A.M., Siu, S., Fiore, N.T., Gerke-Duncan, M.B., Ollerenshaw, S.L., Leigh, S.J., Kunjan, P.A., Kang, J.W., Keay, K.A., 2015. Evidence for a distinct neuro-immune signature in rats that develop behavioural disability after nerve injury. Journal of neuroinflammation 12, 96. Austin, P.J., Beyer, K., Bembrick, A.L., Keay, K.A., 2010. Peripheral nerve injury differentially regulates dopaminergic pathways in the nucleus accumbens of rats with either 'pain alone' or 'pain and disability'. Neuroscience 171, 329-343. Austin, P.J., Moalem-Taylor, G., 2010. The neuro-immune balance in neuropathic pain: Involvement of inflammatory immune cells, immune-like glial cells and cytokines. Journal of Neuroimmunology 229, 26-50. Austin, P.J., Moalem-Taylor, G., 2013. Pathophysiology of neuropathic pain: inflammatory mediators. In: Toth, C., Moulin, D.E. (Eds.), The Condition of Neuropathic Pain. Cambridge University Press, Cambridge University, pp. 77-89. Backonja, M., Coe, C.L., Muller, D.A., Schell, K., 2008. Altered cytokine levels in blood and cerebrospinal fluid of chronic pain patients. Journal of Neuroimmunology 195, 157-163.

Baliki, M.N., Chang, P.C., Baria, A.T., Centeno, M.V., Apkarian, A.V., 2014. Resting-sate functional reorganization of the rat limbic system following neuropathic injury. Scientific reports 4, 6186. Ban, E., Haour, F., Lenstra, R., 1992. Brain interleukin 1 gene expression induced by peripheral lipopolysaccharide administration. Cytokine 4, 48-54. Ban, E., Milon, G., Prudhomme, N., Fillion, G., Haour, F., 1991. Receptors for interleukin-1 (α and β) in mouse brain: Mapping and neuronal localization in hippocampus. Neuroscience 43, 21-30. Bandler, R., Shipley, M.T., 1994. Columnar organization in the midbrain periaqueductal gray: modules for emotional expression? Trends in neurosciences 17, 379-389. Banisadr, G., Fontanges, P., Haour, F., Kitabgi, P., Rostène, W., Mélik Parsadaniantz, S., 2002a. Neuroanatomical distribution of CXCR4 in adult rat brain and its localization in cholinergic and dopaminergic neurons. European Journal of Neuroscience 16, 1661-1671. Banisadr, G., Quéraud-Lesaux, F., Boutterin, M.C., Pélaprat, D., Zalc, B., Rostène, W., Haour, F., Mélik Parsadaniantz, S., 2002b. Distribution, cellular localization and functional role of CCR2 chemokine receptors in adult rat brain. Journal of neurochemistry 81, 257-269. Banks, W.A., 2015. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain, behavior, and immunity 44, 1-8. Banks, W.A., Kastin, A.J., Gutierrez, E.G., 1994. Penetration of interleukin-6 across the murine blood-brain barrier. Neuroscience letters 179, 53-56. Banks, W.A., Ortiz, L., Plotkin, S.R., Kastin, A.J., 1991. Human interleukin (IL) 1 alpha, murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in the mouse by a shared saturable mechanism. The Journal of pharmacology and experimental therapeutics 259, 988-996. Beattie, E.C., Stellwagen, D., Morishita, W., Bresnahan, J.C., Ha, B.K., Von Zastrow, M., Beattie, M.S., Malenka, R.C., 2002. Control of synaptic strength by glial TNFalpha. Science 295, 2282-2285. Bennett, G.J., Xie, Y.K., 1988. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33, 87-107. Bertolucci, P.H.F., de Oliveira, F.F., 2013. Cognitive Impairment in Fibromyalgia. Curr Pain Headache Rep 17, 1-9. Besedovsky, H.O., del Rey, A., 1996. Immune-neuro-endocrine interactions: facts and hypotheses. Endocrine reviews 17, 64-102. Bezzi, P., Domercq, M., Brambilla, L., Galli, R., Schols, D., De Clercq, E., Vescovi, A., Bagetta, G., Kollias, G., Meldolesi, J., Volterra, A., 2001. CXCR4-activated astrocyte glutamate release via TNFalpha: amplification by microglia triggers neurotoxicity. Nature neuroscience 4, 702-710. Biesmans, S., Meert, T.F., Bouwknecht, J.A., Acton, P.D., Davoodi, N., De Haes, P., Kuijlaars, J., Langlois, X., Matthews, L.J., Ver Donck, L., Hellings, N., Nuydens, R., 2013. Systemic immune activation leads to neuroinflammation and sickness behavior in mice. Mediators of inflammation 2013, 271359. Bluthe, R.M., Castanon, N., Pousset, F., Bristow, A., Ball, C., Lestage, J., Michaud, B., Kelley, K.W., Dantzer, R., 1999. Central injection of IL-10 antagonizes the behavioural effects of lipopolysaccharide in rats. Psychoneuroendocrinology 24, 301311. Boelen, A., Kwakkel, J., Thijssen-Timmer, D., Alkemade, A., Fliers, E., Wiersinga, W., 2004. Simultaneous changes in central and peripheral components of the

32

hypothalamus-pituitary-thyroid axis in lipopolysaccharide-induced acute illness in mice. Journal of Endocrinology 182, 315-323. Boelen, A., Kwakkel, J., Wiersinga, W.M., Fliers, E., 2006. Chronic local inflammation in mice results in decreased TRH and type 3 deiodinase mRNA expression in the hypothalamic paraventricular nucleus independently of diminished food intake. Journal of Endocrinology 191, 707-714. Bomholt, S.F., Mikkelsen, J.D., Blackburn-Munro, G., 2005. Normal hypothalamopituitary-adrenal axis function in a rat model of peripheral neuropathic pain. Brain research 1044, 216-226. Brady, L.S., Lynn, A.B., Herkenham, M., Gottesfeld, Z., 1994. Systemic interleukin-1 induces early and late patterns of c-fos mRNA expression in brain. The Journal of neuroscience : the official journal of the Society for Neuroscience 14, 4951-4964. Bramson, C., Herrmann, D.N., Carey, W., Keller, D., Brown, M.T., West, C.R., Verburg, K.M., Dyck, P.J., 2015. Exploring the Role of Tanezumab as a Novel Treatment for the Relief of Neuropathic Pain. Pain medicine 16, 1163-1176. Brischoux, F., Chakraborty, S., Brierley, D.I., Ungless, M.A., 2009. Phasic excitation of dopamine neurons in ventral VTA by noxious stimuli. Proceedings of the National Academy of Sciences of the United States of America 106, 4894-4899. Burke, N.N., Geoghegan, E., Kerr, D.M., Moriarty, O., Finn, D.P., Roche, M., 2013a. Altered neuropathic pain behaviour in a rat model of depression is associated with changes in inflammatory gene expression in the amygdala. Genes, brain, and behavior 12, 705-713. Burke, N.N., Kerr, D.M., Moriarty, O., Finn, D.P., Roche, M., 2014. Minocycline modulates neuropathic pain behaviour and cortical M1-M2 microglial gene expression in a rat model of depression. Brain, behavior, and immunity. Burke, N.N., Llorente, R., Marco, E.M., Tong, K., Finn, D.P., Viveros, M.-P., Roche, M., 2013b. Maternal Deprivation Is Associated With Sex-Dependent Alterations in Nociceptive Behaviour and Neuroinflammatory Mediators in the Rat Following Peripheral Nerve Injury. Pain, 1-12. Butler, M.P., O'Connor, J.J., Moynagh, P.N., 2004. Dissection of tumor-necrosis factor-α inhibition of long-term potentiation (LTP) reveals a p38 mitogen-activated protein kinase-dependent mechanism which maps to early—but not late—phase LTP. Neuroscience 124, 319-326. Cacci, E., Claasen, J.-H., Kokaia, Z., 2005. Microglia-derived tumor necrosis factor-α exaggerates death of newborn hippocampal progenitor cells in vitro. Journal of Neuroscience Research 80, 789-797. Cai, H., Haubensak, W., Anthony, T.E., Anderson, D.J., 2014. Central amygdala PKC-delta(+) neurons mediate the influence of multiple anorexigenic signals. Nature neuroscience 17, 1240-1248. Cao, L., DeLeo, J.A., 2008. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur J Immunol 38, 448458. Capuron, L., Gumnick, J.F., Musselman, D.L., Lawson, D.H., Reemsnyder, A., Nemeroff, C.B., Miller, A.H., 2002. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 26, 643-652. Capuron, L., Miller, A.H., 2011. Immune system to brain signalling: Neuropsychopharmacological implications. Pharmacology & therapeutics 130, 226238.

33

Capuron, L., Pagnoni, G., Demetrashvili, M., Woolwine, B.J., Nemeroff, C.B., Berns, G.S., Miller, A.H., 2005. Anterior cingulate activation and error processing during interferon-alpha treatment. Biological psychiatry 58, 190-196. Capuron, L., Pagnoni, G., Demetrashvili, M.F., Lawson, D.H., Fornwalt, F.B., Woolwine, B., Berns, G.S., Nemeroff, C.B., Miller, A.H., 2007. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 32, 2384-2392. Cardinal, R.N., Parkinson, J.A., Marbini, H.D., Toner, A.J., Bussey, T.J., Robbins, T.W., Everitt, B.J., 2003. Role of the anterior cingulate cortex in the control over behavior by Pavlovian conditioned stimuli in rats. Behavioral neuroscience 117, 566587. Cardoso-Cruz, H., Lima, D., Galhardo, V., 2011. Instability of spatial encoding by CA1 hippocampal place cells after peripheral nerve injury. European Journal of Neuroscience 33, 2255-2264. Cardoso-Cruz, H., Lima, D., Galhardo, V., 2013. Impaired spatial memory performance in a rat model of neuropathic pain is associated with reduced hippocampus-prefrontal cortex connectivity. Journal of Neuroscience 33, 2465-2480. Castanon, N., Médina, C., Mormède, C., Dantzer, R., 2004. Chronic administration of tianeptine balances lipopolysaccharide-induced expression of cytokines in the spleen and hypothalamus of rats. Psychoneuroendocrinology 29, 778-790. Chang, P.C., Pollema-Mays, S.L., Centeno, M.V., Procissi, D., Contini, M., Baria, A.T., Martina, M., Apkarian, A.V., 2014. Role of nucleus accumbens in neuropathic pain: linked multi-scale evidence in the rat transitioning to neuropathic pain. Pain 155, 1128-1139. Chiang, R.P., Huang, C.T., Tsai, Y.J., 2013. Melatonin reduces median nerve injuryinduced mechanical hypersensitivity via inhibition of microglial p38 mitogenactivated protein kinase activation in rat cuneate nucleus. Journal of pineal research 54, 232-244. Chou, C.W., Wong, G.T., Lim, G., McCabe, M.F., Wang, S., Irwin, M.G., Mao, J., 2011. Peripheral nerve injury alters the expression of NF-kappaB in the rat's hippocampus. Brain research 1378, 66-71. Chu, H., Sun, J., Xu, H., Niu, Z., Xu, M., 2012. Effect of periaqueductal gray melanocortin 4 receptor in pain facilitation and glial activation in rat model of chronic constriction injury. Neurological research 34, 871-888. Costigan, M., Moss, A., Latremoliere, A., Johnston, C., Verma-Gandhu, M., Herbert, T.A., Barrett, L., Brenner, G.J., Vardeh, D., Woolf, C.J., Fitzgerald, M., 2009a. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. The Journal of neuroscience : the official journal of the Society for Neuroscience 29, 14415-14422. Costigan, M., Scholz, J., Woolf, C.J., 2009b. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci 32, 1-32. Coull, J.A.M., Beggs, S., Boudreau, D., Boivin, D., Tsuda, M., Inoue, K., Gravel, C., Salter, M.W., De Koninck, Y., 2005. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 438, 1017-1021. Covey, W.C., Ignatowski, T.A., Knight, P.R., Spengler, R.N., 2000. Brain-derived TNFalpha: involvement in neuroplastic changes implicated in the conscious perception of persistent pain. Brain research 859, 113-122. Craig, A.D., 2003. Interoception: the sense of the physiological condition of the body. Current opinion in neurobiology 13, 500-505.

34

Cunningham, A.J., Murray, C.A., O'Neill, L.A., Lynch, M.A., O'Connor, J.J., 1996. Interleukin-1 beta (IL-1 beta) and tumour necrosis factor (TNF) inhibit long-term potentiation in the rat dentate gyrus in vitro. Neuroscience letters 203, 17-20. D'Mello, C., Le, T., Swain, M.G., 2009. Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation. The Journal of neuroscience : the official journal of the Society for Neuroscience 29, 2089-2102. D'Mello, C., Riazi, K., Le, T., Stevens, K.M., Wang, A., McKay, D.M., Pittman, Q.J., Swain, M.G., 2013. P-selectin-mediated monocyte-cerebral endothelium adhesive interactions link peripheral organ inflammation to sickness behaviors. The Journal of neuroscience : the official journal of the Society for Neuroscience 33, 14878-14888. Dantzer, R., 2009. Cytokine, Sickness Behaviour and Depression. Immunol Allergy Clin N AM 29, 247-264. Dantzer, R., O'Connor, J.C., Freund, G.G., Johnson, R.W., Kelley, K.W., 2008. From inflammation to sickness and depression: when the immune system subjugates the brain. Nature reviews. Neuroscience 9, 46-56. Davis, D.A., Luecken, L.J., Zautra, A.J., 2005. Are reports of childhood abuse related to the experience of chronic pain in adulthood? A meta-analytic review of the literature. The Clinical journal of pain 21, 398-405. de Novellis, V., Vita, D., Gatta, L., Luongo, L., Bellini, G., De Chiaro, M., Marabese, I., Siniscalco, D., Boccella, S., Piscitelli, F., Di Marzo, V., Palazzo, E., Rossi, F., Maione, S., 2011. The blockade of the transient receptor potential vanilloid type 1 and fatty acid amide hydrolase decreases symptoms and central sequelae in the medial prefrontal cortex of neuropathic rats. Molecular pain 7, 7. Dean, O.M., Data-Franco, J., Giorlando, F., Berk, M., 2012. Minocycline: therapeutic potential in psychiatry. CNS drugs 26, 391-401. Decosterd, I., Woolf, C.J., 2000. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87, 149-158. del Rey, A., Yau, H.-J., Randolf, A., Centeno, M.V., Wildmann, J., Martina, M., Besedovsky, H.O., Apkarian, A.V., 2011. Chronic neuropathic pain-like behaviour correlates with IL-1beta expression and disrupts cytokine interactions in the hippocampus. Pain 152, 2827-2835. Dellarole, A., Morton, P., Brambilla, R., Walters, W., Summers, S., Bernardes, D., Grilli, M., Bethea, J.R., 2014. Neuropathic pain-induced depressive-like behavior and hippocampal neurogenesis and plasticity are dependent on TNFR1 signaling. Brain, behavior, and immunity 41, 65-81. Dick, B.D., Rashiq, S., 2007. Disruption of attention and working memory traces in individuals with chronic pain. Anesthesia and analgesia 104, 1223-1229. Dimitrov, E.L., Tsuda, M.C., Cameron, H.A., Usdin, T.B., 2014. Anxiety- and depression-like behavior and impaired neurogenesis evoked by peripheral neuropathy persist following resolution of prolonged tactile hypersensitivity. The Journal of neuroscience : the official journal of the Society for Neuroscience 34, 12304-12312. Dowlati, Y., Herrmann, N., Swardfager, W., Liu, H., Sham, L., Reim, E.K., Lanctot, K.L., 2010. A meta-analysis of cytokines in major depression. Biological psychiatry 67, 446-457. Duric, V., McCarson, K.E., 2005. Hippocampal neurokinin-1 receptor and brainderived neurotrophic factor gene expression is decreased in rat models of pain and stress. Neuroscience 133, 999-1006. Duric, V., McCarson, K.E., 2006. Persistent Pain Produces Stress-like Alterations in Hippocampal Neurogenesis and Gene Expression. The Journal of Pain 7, 544-555.

35

Duric, V., McCarson, K.E., 2007. Neurokinin-1 (NK-1) receptor and brain-derived neurotrophic factor (BDNF) gene expression is differentially modulated in the rat spinal dorsal horn and hippocampus during inflammatory pain. Molecular pain 3, 32. Dutar, P., Lamour, Y., Jobert, A., 1985. Activation of identified septo-hippocampal neurons by noxious peripheral stimulation. Brain research 328, 15-21. Echeverry, S., Shi, X.Q., Rivest, S., Zhang, J., 2011. Peripheral nerve injury alters blood-spinal cord barrier functional and molecular integrity through a selective inflammatory pathway. The Journal of neuroscience : the official journal of the Society for Neuroscience 31, 10819-10828. Ewan, E.E., Martin, T.J., 2011a. Opioid facilitation of rewarding electrical brain stimulation is suppressed in rats with neuropathic pain. Anesthesiology 114, 624-632. Ewan, E.E., Martin, T.J., 2011b. Rewarding electrical brain stimulation in rats after peripheral nerve injury: decreased facilitation by commonly abused prescription opioids. Anesthesiology 115, 1271-1280. Fanselow, M.S., Dong, H.-W., 2010. Are the Dorsal and Ventral Hippocampus Functionally Distinct Structures. Neuron 65, 7-19. Fanselow, M.S., Poulos, A.M., 2005. The neuroscience of mammalian associative learning. Annual review of psychology 56, 207-234. Farrar, W.L., Kilian, P.L., Ruff, M.R., Hill, J.M., Pert, C.B., 1987. Visualization and characterization of interleukin 1 receptors in brain. The Journal of Immunology 139, 459-463. Fishbain, D.A., Cutler, R., Rosomoff, H.L., Rosomoff, R.S., 1997. Chronic painassociated depression: antecedent or consequence of chronic pain? A review. The Clinical journal of pain 13, 116-137. Fleming, J.C., Bao, F., Chen, Y., Hamilton, E.F., Gonzalez-Lara, L.E., Foster, P.J., Weaver, L.C., 2009. Timing and duration of anti-alpha4beta1 integrin treatment after spinal cord injury: effect on therapeutic efficacy. J Neurosurg Spine 11, 575-587. Fuccio, C., Luongo, C., Capodanno, P., Giordano, C., Scafuro, M.A., Siniscalco, D., Lettieri, B., Rossi, F., Maione, S., Berrino, L., 2009. A single subcutaneous injection of ozone prevents allodynia and decreases the over-expression of pro-inflammatory caspases in the orbito-frontal cortex of neuropathic mice. European journal of pharmacology 603, 42-49. Fukuhara, K., Ishikawa, K., Yasuda, S., Kishishita, Y., Kim, H.K., Kakeda, T., Yamamoto, M., Norii, T., Ishikawa, T., 2012. Intracerebroventricular 4methylcatechol (4-MC) ameliorates chronic pain associated with depression-like behavior via induction of brain-derived neurotrophic factor (BDNF). Cellular and molecular neurobiology 32, 971-977. Furukawa, K., Mattson, M.P., 1998. The Transcription Factor NF-κB Mediates Increases in Calcium Currents and Decreases in NMDA- and AMPA/Kainate-Induced Currents Induced by Tumor Necrosis Factor-α in Hippocampal Neurons. Journal of Neurochemistry 70, 1876-1886. Gabellec, M.M., Griffais, R., Fillion, G., Haour, F., 1995. Expression of interleukin 1 alpha, interleukin 1 beta and interleukin 1 receptor antagonist mRNA in mouse brain: regulation by bacterial lipopolysaccharide (LPS) treatment. Brain research. Molecular brain research 31, 122-130. Gadient, R.A., Otten, U., 1993. Differential expression of interleukin-6 (IL-6) and interleukin-6 receptor (IL-6R) mRNAs in rat hypothalamus. Neuroscience letters 153, 13-16.

36

Gadient, R.A., Otten, U., 1994. Identification of interleukin-6 (IL-6)-expressing neurons in the cerebellum and hippocampus of normal adult rats. Neuroscience letters 182, 243-246. Gerard, E., Spengler, R.N., Bonoiu, A.C., Mahajan, S.D., Davidson, B.A., Ding, H., Kumar, R., Prasad, P.N., Knight, P.R., Ignatowski, T.A., 2015. Chronic Constriction Injury-Induced Nociception Is Relieved by Nanomedicine-Mediated Decrease of Rat Hippocampal Tumor Necrosis Factor. Pain. Gerashchenko, D., Blanco-Centurion, C., Greco, M.A., Shiromani, P.J., 2003. Effects of lateral hypothalamic lesion with the neurotoxin hypocretin-2-saporin on sleep in Long-Evans rats. Neuroscience 116, 223-235. Gerashchenko, D., Kohls, M.D., Greco, M., Waleh, N.S., Salin-Pascual, R., Kilduff, T.S., Lappi, D.A., Shiromani, P.J., 2001. Hypocretin-2-saporin lesions of the lateral hypothalamus produce narcoleptic-like sleep behavior in the rat. The Journal of neuroscience : the official journal of the Society for Neuroscience 21, 7273-7283. Gerashchenko, D., Shiromani, P.J., 2004. Different neuronal phenotypes in the lateral hypothalamus and their role in sleep and wakefulness. Molecular neurobiology 29, 41-59. Giordano, C., Cristino, L., Luongo, L., Siniscalco, D., Petrosino, S., Piscitelli, F., Marabese, I., Gatta, L., Rossi, F., Imperatore, R., Palazzo, E., de Novellis, V., Di Marzo, V., Maione, S., 2012. TRPV1-dependent and -independent alterations in the limbic cortex of neuropathic mice: impact on glial caspases and pain perception. Cerebral cortex 22, 2495-2518. Goehler, L.E., Gaykema, R.P., Nguyen, K.T., Lee, J.E., Tilders, F.J., Maier, S.F., Watkins, L.R., 1999. Interleukin-1beta in immune cells of the abdominal vagus nerve: a link between the immune and nervous systems? The Journal of neuroscience : the official journal of the Society for Neuroscience 19, 2799-2806. Goffer, Y., Xu, D., Eberle, S.E., D'Amour, J., Lee, M., Tukey, D., Froemke, R.C., Ziff, E.B., Wang, J., 2013. Calcium-permeable AMPA receptors in the nucleus accumbens regulate depression-like behaviors in the chronic neuropathic pain state. The Journal of neuroscience : the official journal of the Society for Neuroscience 33, 19034-19044. Goins, W.F., Cohen, J.B., Glorioso, J.C., 2012. Gene therapy for the treatment of chronic peripheral nervous system pain. Neurobiol Dis 48, 255-270. Goncalves, L., Silva, R., Pinto-Ribeiro, F., Pego, J.M., Bessa, J.M., Pertovaara, A., Sousa, N., Almeida, A., 2008. Neuropathic pain is associated with depressive behaviour and induces neuroplasticity in the amygdala of the rat. Experimental neurology 213, 48-56. Goshen, I., Kreisel, T., Ben-Menachem-Zidon, O., Licht, T., Weidenfeld, J., Ben-Hur, T., Yirmiya, R., 2008. Brain interleukin-1 mediates chronic stress-induced depression in mice via adrenocortical activation and hippocampal neurogenesis suppression. Molecular psychiatry 13, 717-728. Goshen, I., Kreisel, T., Ounallah-Saad, H., Renbaum, P., Zalzstein, Y., Ben-Hur, T., Levy-Lahad, E., Yirmiya, R., 2007. A dual role for interleukin-1 in hippocampaldependent memory processes. Psychoneuroendocrinology 32, 1106-1115. Gosselin, R.D., Bebber, D., Decosterd, I., 2010. Upregulation of the GABA transporter GAT-1 in the gracile nucleus in the spared nerve injury model of neuropathic pain. Neuroscience letters 480, 132-137. Grace, P.M., Hutchinson, M.R., Maier, S.F., Watkins, L.R., 2014. Pathological pain and the neuroimmune interface. Nat Rev Immunol 14, 217-231.

37

Gregoire, S., Michaud, V., Chapuy, E., Eschalier, A., Ardid, D., 2012. Study of emotional and cognitive impairments in mononeuropathic rats: effect of duloxetine and gabapentin. Pain 153, 1657-1663. Guillemin, G.J., Kerr, S.J., Smythe, G.A., Smith, D.G., Kapoor, V., Armati, P.J., Croitoru, J., Brew, B.J., 2001. Kynurenine pathway metabolism in human astrocytes: a paradox for neuronal protection. Journal of neurochemistry 78, 842-853. Guo, W., Wang, H., Zou, S., Dubner, R., Ren, K., 2012. Chemokine signaling involving chemokine (C-C motif) ligand 2 plays a role in descending pain facilitation. Neuroscience bulletin 28, 193-207. Haas, H.S., Schauenstein, K., 1997. Neuroimmunomodulation via limbic structures-the neuroanatomy of psychoimmunology. Prog Neurobiol 51, 195-222. Hansen, M.K., O'Connor, K.A., Goehler, L.E., Watkins, L.R., Maier, S.F., 2001. The contribution of the vagus nerve in interleukin-1beta-induced fever is dependent on dose. American journal of physiology. Regulatory, integrative and comparative physiology 280, R929-934. Hart, R.P., Martelli, M.F., Zasler, N.D., 2000. Chronic pain and neuropsychological functioning. Neuropsychology Review 10, 131-149. Haydon, P.G., Carmignoto, G., 2006. Astrocyte control of synaptic transmission and neurovascular coupling. Physiological reviews 86, 1009-1031. Henry, C.J., Huang, Y., Wynne, A., Hanke, M., Himler, J., Bailey, M.T., Sheridan, J.F., Godbout, J.P., 2008. Minocycline attenuates lipopolysaccharide (LPS)-induced neuroinflammation, sickness behavior, and anhedonia. Journal of neuroinflammation 5, 15. Ho, Y.C., Cheng, J.K., Chiou, L.C., 2013. Hypofunction of glutamatergic neurotransmission in the periaqueductal gray contributes to nerve-injury-induced neuropathic pain. The Journal of neuroscience : the official journal of the Society for Neuroscience 33, 7825-7836. Horuk, R., Martin, A.W., Wang, Z., Schweitzer, L., Gerassimides, A., Guo, H., Lu, Z., Hesselgesser, J., Perez, H.D., Kim, J., Parker, J., Hadley, T.J., Peiper, S.C., 1997. Expression of chemokine receptors by subsets of neurons in the central nervous system. The Journal of Immunology 158, 2882-2890. Hu, Y., Yang, J., Hu, Y., Wang, Y., Li, W., 2010. Amitriptyline rather than lornoxicam ameliorates neuropathic pain-induced deficits in abilities of spatial learning and memory. European journal of anaesthesiology 27, 162-168. Huang, C.T., Chiang, R.P., Chen, C.L., Tsai, Y.J., 2014. Sleep deprivation aggravates median nerve injury-induced neuropathic pain and enhances microglial activation by suppressing melatonin secretion. Sleep 37, 1513-1523. Hutchinson, M.R., Watkins, L.R., 2014. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 76 Pt B, 218-227. Ida, T., Hara, M., Nakamura, Y., Kozaki, S., Tsunoda, S., Ihara, H., 2008. Cytokineinduced enhancement of calcium-dependent glutamate release from astrocytes mediated by nitric oxide. Neuroscience letters 432, 232-236. Ignatowski, T.A., Covey, W.C., Knight, P.R., Severin, C.M., Nickola, T.J., Spengler, R.N., 1999. Brain-derived TNFalpha mediates neuropathic pain. Brain research 841, 70-77. Ignatowski, T.A., Sud, R., Reynolds, J.L., Knight, P.R., Spengler, R.N., 2005. The dissipation of neuropathic pain paradoxically involves the presence of tumor necrosis factor-α (TNF). Neuropharmacology 48, 448-460. Iliff, J.J., Wang, M., Liao, Y., Plogg, B.A., Peng, W., Gundersen, G.A., Benveniste, H., Vates, G.E., Deane, R., Goldman, S.A., Nagelhus, E.A., Nedergaard, M., 2012. A

38

Paravascular Pathway Facilitates CSF Flow Through the Brain Parenchyma and the Clearance of Interstitial Solutes, Including Amyloid β. Janak, P.H., Tye, K.M., 2015. From circuits to behaviour in the amygdala. Nature 517, 284-292. Jennings, J.H., Sparta, D.R., Stamatakis, A.M., Ung, R.L., Pleil, K.E., Kash, T.L., Stuber, G.D., 2013. Distinct extended amygdala circuits for divergent motivational states. Nature 496, 224-228. Jensen, T.S., Finnerup, N.B., 2007. Management of neuropathic pain. Current opinion in supportive and palliative care 1, 126-131. Johansen, J.P., Fields, H.L., 2004. Glutamatergic activation of anterior cingulate cortex produces an aversive teaching signal. Nature neuroscience 7, 398-403. Kakucska, I., Romero, L.I., Clark, B.D., Rondeel, J.M., Qi, Y., Alex, S., Emerson, C.H., Lechan, R.M., 1994. Suppression of thyrotropin-releasing hormone gene expression by interleukin-1-beta in the rat: implications for nonthyroidal illness. Neuroendocrinology 59, 129-137. Kalliomaki, J., Attal, N., Jonzon, B., Bach, F.W., Huizar, K., Ratcliffe, S., Eriksson, B., Janecki, M., Danilov, A., Bouhassira, D., Group, A.P.S., 2013. A randomized, double-blind, placebo-controlled trial of a chemokine receptor 2 (CCR2) antagonist in posttraumatic neuralgia. Pain 154, 761-767. Kalman, E., Keay, K.A., 2014. Different patterns of morphological changes in the hippocampus and dentate gyrus accompany the differential expression of disability following nerve injury. Journal of anatomy 225, 591-603. Keay, K.A., Bandler, R., 2001. Parallel circuits mediating distinct emotional coping reactions to different types of stress. Neuroscience and biobehavioral reviews 25, 669678. Keay, K.A., Monassi, C.R., Levison, D.B., Bandler, R., 2004. Peripheral nerve injury evokes disabilities and sensory dysfunction in a subpopulation of rats: a closer model to human chronic neuropathic pain? Neuroscience letters 361, 188-191. Kehlet, H., Jensen, T.S., Woolf, C.J., 2006. Persistent postsurgical pain: risk factors and prevention. Lancet 367, 1618-1625. Khanna, S., Sinclair, J.G., 1989. Noxious stimuli produce prolonged changes in the CA1 region of the rat hippocampus. PAIN 39, 337-343. Kilburn-Watt, E., Banati, R.B., Keay, K.A., 2010. Altered thyroid hormones and behavioural change in a sub-population of rats following chronic constriction injury. Journal of neuroendocrinology 22, 960-970. Kilburn-Watt, E., Banati, R.B., Keay, K.A., 2014. Rats with altered behaviour following nerve injury show evidence of centrally altered thyroid regulation. Brain research bulletin 107, 110-118. Kim, H., Chen, L., Lim, G., Sung, B., Wang, S., McCabe, M.F., Rusanescu, G., Yang, L., Tian, Y., Mao, J., 2012. Brain indoleamine 2,3-dioxygenase contributes to the comorbidity of pain and depression. The Journal of clinical investigation 122, 29402954. Kim, H.W., Chang, Y.C., Chen, M., Rapoport, S.I., Rao, J.S., 2009. Chronic NMDA administration to rats increases brain pro-apoptotic factors while decreasing antiApoptotic factors and causes cell death. BMC neuroscience 10, 123. Kim, K.J., Yoon, Y.W., Chung, J.M., 1997. Comparison of three rodent neuropathic pain models. Exp Brain Res 113, 200-206. Kim, S.H., Chung, J.M., 1992. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 50, 355-363.

39

Kim, S.Y., Adhikari, A., Lee, S.Y., Marshel, J.H., Kim, C.K., Mallory, C.S., Lo, M., Pak, S., Mattis, J., Lim, B.K., Malenka, R.C., Warden, M.R., Neve, R., Tye, K.M., Deisseroth, K., 2013. Diverging neural pathways assemble a behavioural state from separable features in anxiety. Nature 496, 219-223. Kinouchi, K., Brown, G., Pasternak, G., Donner, D.B., 1991. Identification and characterization of receptors for tumor necrosis factor-α in the brain. Biochemical and biophysical research communications 181, 1532-1538. Knezevic, N.N., Cicmil, N., Knezevic, I., Candido, K.D., 2015. Discontinued neuropathic pain therapy between 2009-2015. Expert opinion on investigational drugs 24, 1631-1646. Knobloch, H.S., Charlet, A., Hoffmann, L.C., Eliava, M., Khrulev, S., Cetin, A.H., Osten, P., Schwarz, M.K., Seeburg, P.H., Stoop, R., Grinevich, V., 2012. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 73, 553-566. Kodama, D., Ono, H., Tanabe, M., 2007. Altered hippocampal long-term potentiation after peripheral nerve injury in mice. European journal of pharmacology 574, 127132. Kodama, D., Ono, H., Tanabe, M., 2011. Increased hippocampal glycine uptake and cognitive dysfunction after peripheral nerve injury. Pain 152, 809-817. Konsman, J.P., Kelley, K., Dantzer, R., 1999. Temporal and spatial relationships between lipopolysaccharide-induced expression of Fos, interleukin-1beta and inducible nitric oxide synthase in rat brain. Neuroscience 89, 535-548. Konsman, J.P., Parnet, P., Dantzer, R., 2002. Cytokine-induced sickness behaviour: mechanisms and implications. Trends in neurosciences 25, 154-159. Koo, J.W., Duman, R.S., 2008. IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress. Proceedings of the National Academy of Sciences of the United States of America 105, 751-756. Kwilasz, A.J., Grace, P.M., Serbedzija, P., Maier, S.F., Watkins, L.R., 2015. The therapeutic potential of interleukin-10 in neuroimmune diseases. Neuropharmacology 96, 55-69. LaBuda, C.J., Fuchs, P.N., 2005. Attenuation of negative pain affect produced by unilateral spinal nerve injury in the rat following anterior cingulate cortex activation. Neuroscience 136, 311-322. LaGraize, S.C., Labuda, C.J., Rutledge, M.A., Jackson, R.L., Fuchs, P.N., 2004. Differential effect of anterior cingulate cortex lesion on mechanical hypersensitivity and escape/avoidance behavior in an animal model of neuropathic pain. Experimental neurology 188, 139-148. Lammel, S., Ion, D.I., Roeper, J., Malenka, R.C., 2011. Projection-specific modulation of dopamine neuron synapses by aversive and rewarding stimuli. Neuron 70, 855-862. Lammel, S., Lim, B.K., Malenka, R.C., 2014. Reward and aversion in a heterogeneous midbrain dopamine system. Neuropharmacology 76 Pt B, 351-359. Lammel, S., Lim, B.K., Ran, C., Huang, K.W., Betley, M.J., Tye, K.M., Deisseroth, K., Malenka, R.C., 2012. Input-specific control of reward and aversion in the ventral tegmental area. Nature 491, 212-217. Landry, R.P., Jacobs, V.L., Romero-Sandoval, E.A., DeLeo, J.A., 2012. Propentofylline, a CNS glial modulator does not decrease pain in post-herpetic neuralgia patients: in vitro evidence for differential responses in human and rodent microglia and macrophages. Experimental neurology 234, 340-350.

40

Laye, S., Gheusi, G., Cremona, S., Combe, C., Kelley, K., Dantzer, R., Parnet, P., 2000. Endogenous brain IL-1 mediates LPS-induced anorexia and hypothalamic cytokine expression. American journal of physiology. Regulatory, integrative and comparative physiology 279, R93-98. Laye, S., Parnet, P., Goujon, E., Dantzer, R., 1994. Peripheral administration of lipopolysaccharide induces the expression of cytokine transcripts in the brain and pituitary of mice. Brain research. Molecular brain research 27, 157-162. LeBlanc, B.W., Zerah, M.L., Kadasi, L.M., Chai, N., Saab, C.Y., 2011. Minocycline injection in the ventral posterolateral thalamus reverses microglial reactivity and thermal hyperalgesia secondary to sciatic neuropathy. Neurosci Lett 498, 138-142. Ledeboer, A., Gamanos, M., Lai, W., Martin, D., Maier, S.F., Watkins, L.R., Quan, N., 2005. Involvement of spinal cord nuclear factor kappaB activation in rat models of proinflammatory cytokine-mediated pain facilitation. Eur J Neurosci 22, 1977-1986. Leite-Almeida, H., Cerqueira, J.J., Wei, H., Ribeiro-Costa, N., Anjos-Martins, H., Sousa, N., Pertovaara, A., Almeida, A., 2012. Differential effects of left/right neuropathy on rats' anxiety and cognitive behavior. Pain 153, 2218-2225. Li, X., Wang, J., Wang, Z., Dong, C., Dong, X., Jing, Y., Yuan, Y., Fan, G., 2008. Tumor necrosis factor-alpha of Red nucleus involved in the development of neuropathic allodynia. Brain research bulletin 77, 233-236. Liu, J., Feng, X., Yu, M., Xie, W., Zhao, X., Li, W., Guan, R., Xu, J., 2007. Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain. Neuroscience letters 412, 268-272. Liu, L., Tornqvist, E., Mattsson, P., Eriksson, N.P., Persson, J.K., Morgan, B.P., Aldskogius, H., Svensson, M., 1995. Complement and clusterin in the spinal cord dorsal horn and gracile nucleus following sciatic nerve injury in the adult rat. Neuroscience 68, 167-179. Liu, M.G., Chen, J., 2014. Preclinical research on pain comorbidity with affective disorders and cognitive deficits: Challenges and perspectives. Prog Neurobiol 116, 13-32. Liu, Y., Ho, R.C., Mak, A., 2012. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. Journal of affective disorders 139, 230-239. Loggia, M.L., Chonde, D.B., Akeju, O., Arabasz, G., Catana, C., Edwards, R.R., Hill, E., Hsu, S., Izquierdo-Garcia, D., Ji, R.R., Riley, M., Wasan, A.D., Zurcher, N.R., Albrecht, D.S., Vangel, M.G., Rosen, B.R., Napadow, V., Hooker, J.M., 2015. Evidence for brain glial activation in chronic pain patients. Brain : a journal of neurology 138, 604-615. Louveau, A., Smirnov, I., Keyes, T.J., Eccles, J.D., Rouhani, S.J., Peske, J.D., Derecki, N.C., Castle, D., Mandell, J.W., Lee, K.S., Harris, T.H., Kipnis, J., 2015. Structural and functional features of central nervous system lymphatic vessels. Nature. Lu, J., Jhou, T.C., Saper, C.B., 2006. Identification of wake-active dopaminergic neurons in the ventral periaqueductal gray matter. The Journal of neuroscience : the official journal of the Society for Neuroscience 26, 193-202. Majd, M., Hashemian, F., Hosseini, S.M., Vahdat Shariatpanahi, M., Sharifi, A., 2015. A Randomized, Double-blind, Placebo-controlled Trial of Celecoxib Augmentation of Sertraline in Treatment of Drug-naive Depressed Women: A Pilot Study. Iranian Journal of Pharmaceutical Research : IJPR 14, 891-899.

41

Marcello, L., Cavaliere, C., Colangelo, A.M., Bianco, M.R., Cirillo, G., Alberghina, L., Papa, M., 2013. Remodelling of supraspinal neuroglial network in neuropathic pain is featured by a reactive gliosis of the nociceptive amygdala. European journal of pain 17, 799-810. Martin, T.J., Kim, S.A., Buechler, N.L., Porreca, F., Eisenach, J.C., 2007. Opioid selfadministration in the nerve-injured rat: relevance of antiallodynic effects to drug consumption and effects of intrathecal analgesics. Anesthesiology 106, 312-322. Martini, R., Willison, H., 2016. Neuroinflammation in the peripheral nerve: Cause, modulator, or bystander in peripheral neuropathies? Glia 64, 475-486. McAfoose, J., Baune, B.T., 2009. Evidence for a cytokine model of cognitive function. Neuroscience and biobehavioral reviews 33, 355-366. Menefee, L.A., Frank, E.D., Doghramji, K., Picarello, K., Park, J.J., Jalali, S., PerezSchwartz, L., 2000. Self-reported sleep quality and quality of life for individuals with chronic pain conditions. The Clinical journal of pain 16, 290-297. Metz, A.E., Yau, H.J., Centeno, M.V., Apkarian, A.V., Martina, M., 2009. Morphological and functional reorganization of rat medial prefrontal cortex in neuropathic pain. Proceedings of the National Academy of Sciences of the United States of America 106, 2423-2428. Meyer-Rosberg, K., Kvarnstrom, A., Kinnman, E., Gordh, T., Nordfors, L.O., Kristofferson, A., 2001. Peripheral neuropathic pain--a multidimensional burden for patients. European journal of pain 5, 379-389. Mitchell, K., Yang, H.Y., Berk, J.D., Tran, J.H., Iadarola, M.J., 2009. Monocyte chemoattractant protein-1 in the choroid plexus: a potential link between vascular proinflammatory mediators and the CNS during peripheral tissue inflammation. Neuroscience 158, 885-895. Miyaoka, T., Wake, R., Furuya, M., Liaury, K., Ieda, M., Kawakami, K., Tsuchie, K., Taki, M., Ishihara, K., Araki, T., Horiguchi, J., 2012. Minocycline as adjunctive therapy for patients with unipolar psychotic depression: An open-label study. Progress in Neuro-Psychopharmacology and Biological Psychiatry 37, 222-226. Miyaoka, T., Yasukawa, R., Yasuda, H., Hayashida, M., Inagaki, T., Horiguchi, J., 2008. Minocycline as Adjunctive Therapy for Schizophrenia: An Open-Label Study. Clinical Neuropharmacology 31, 287-292. Monassi, C.R., Bandler, R., Keay, K.A., 2003. A subpopulation of rats show social and sleep-waking changes typical of chronic neuropathic pain following peripheral nerve injury. Eur J Neurosci 17, 1907-1920. Monje, M.L., Toda, H., Palmer, T.D., 2003. Inflammatory Blockade Restores Adult Hippocampal Neurogenesis. Science 302, 1760-1765. Mor, D., Bembrick, A.L., Austin, P.J., Keay, K.A., 2011. Evidence for cellular injury in the midbrain of rats following chronic constriction injury of the sciatic nerve. Journal of chemical neuroanatomy 41, 158-169. Mor, D., Bembrick, A.L., Austin, P.J., Wyllie, P.M., Creber, N.J., Denyer, G.S., Keay, K.A., 2010. Anatomically specific patterns of glial activation in the periaqueductal gray of the sub-population of rats showing pain and disability following chronic constriction injury of the sciatic nerve. Neuroscience 166, 11671184. Mor, D., Keay, K.A., 2013. Differential regulation of glucocorticoid receptor expression in distinct columns of periaqueductal grey in rats with behavioural disability following nerve injury. Cellular and molecular neurobiology 33, 953-963.

42

Moriarty, O., McGuire, B.E., Finn, D.P., 2011. The effect of pain on cognitive function: A review of clinical and preclinical research. Progress in Neurobiology 93, 385-404. Moron, J.A., Zakharova, I., Ferrer, J.V., Merrill, G.A., Hope, B., Lafer, E.M., Lin, Z.C., Wang, J.B., Javitch, J.A., Galli, A., Shippenberg, T.S., 2003. Mitogen-activated protein kinase regulates dopamine transporter surface expression and dopamine transport capacity. The Journal of neuroscience : the official journal of the Society for Neuroscience 23, 8480-8488. Moser, M.B., Moser, E.I., 1998. Functional differentiation in the hippocampus. Hippocampus 8, 608-619. Muller, N., Schwarz, M.J., Dehning, S., Douhe, A., Cerovecki, A., Goldstein-Muller, B., Spellmann, I., Hetzel, G., Maino, K., Kleindienst, N., Moller, H.J., Arolt, V., Riedel, M., 2006. The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Molecular psychiatry 11, 680-684. Murray, E.A., Wise, S.P., Rhodes, S.E.V., 2011. What Can Different Brains Do with Reward? In: Gottfried, J.A. (Ed.), Neurobiology of Sensation and Reward, Boca Raton (FL). Mutso, A.A., Radzicki, D., Baliki, M.N., Centeno, M., Radulovic, J., Martina, M., Miler, R.J., Apkarian, A.V., 2012. Abnormalities in Hippocampal Function with Persistent Pain. The Journal of Neuroscience 32, 5747-5756. Narita, M., Kuzumaki, N., Narita, M., Kaneko, C., Hareyama, N., Miyatake, M., Shindo, K., Miyoshi, K., Nakajima, M., Nagumo, Y., Sato, F., Wachi, H., Seyama, Y., Suzuki, T., 2006. Chronic pain-induced emotional dysfunction is associated with astrogliosis due to cortical delta-opioid receptor dysfunction. Journal of neurochemistry 97, 1369-1378. Narita, M., Niikura, K., Nanjo-Niikura, K., Narita, M., Furuya, M., Yamashita, A., Saeki, M., Matsushima, Y., Imai, S., Shimizu, T., Asato, M., Kuzumaki, N., Okutsu, D., Miyoshi, K., Suzuki, M., Tsukiyama, Y., Konno, M., Yomiya, K., Matoba, M., Suzuki, T., 2011. Sleep disturbances in a neuropathic pain-like condition in the mouse are associated with altered GABAergic transmission in the cingulate cortex. Pain 152, 1358-1372. Nascimento, F.P., Macedo-Junior, S.J., Borges, F.R., Cremonese, R.P., da Silva, M.D., Luiz-Cerutti, M., Martins, D.F., Rodrigues, A.L., Santos, A.R., 2015. Thalidomide reduces mechanical hyperalgesia and depressive-like behavior induced by peripheral nerve crush in mice. Neuroscience 303, 51-58. Neis, V.B., Manosso, L.M., Moretti, M., Freitas, A.E., Daufenbach, J., Rodrigues, A.L., 2014. Depressive-like behavior induced by tumor necrosis factor-alpha is abolished by agmatine administration. Behavioural brain research 261, 336-344. Norman, G.J., Karelina, K., Morris, J.S., Zhang, N., Cochran, M., Courtney DeVries, A., 2010a. Social interaction prevents the development of depressive-like behavior post nerve injury in mice: a potential role for oxytocin. Psychosomatic medicine 72, 519-526. Norman, G.J., Karelina, K., Zhang, N., Walton, J.C., Morris, J.S., Devries, A.C., 2010b. Stress and IL-1beta contribute to the development of depressive-like behavior following peripheral nerve injury. Molecular psychiatry 15, 404-414. O'Connor, J.C., Lawson, M.A., Andre, C., Moreau, M., Lestage, J., Castanon, N., Kelley, K.W., Dantzer, R., 2009. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Molecular psychiatry 14, 511-522.

43

Ongur, D., Price, J.L., 2000. The organization of networks within the orbital and medial prefrontal cortex of rats, monkeys and humans. Cerebral cortex 10, 206-219. Orsini, C.A., Trotta, R.T., Bizon, J.L., Setlow, B., 2015. Dissociable roles for the basolateral amygdala and orbitofrontal cortex in decision-making under risk of punishment. The Journal of neuroscience : the official journal of the Society for Neuroscience 35, 1368-1379. Ozaki, S., Narita, M., Narita, M., Iino, M., Sugita, J., Matsumura, Y., Suzuki, T., 2002. Suppression of the morphine-induced rewarding effect in the rat with neuropathic pain: implication of the reduction in mu-opioid receptor functions in the ventral tegmental area. Journal of neurochemistry 82, 1192-1198. Pace, T.W., Hu, F., Miller, A.H., 2007. Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain, behavior, and immunity 21, 9-19. Pan, W., Kastin, A.J., 2002. TNFalpha transport across the blood-brain barrier is abolished in receptor knockout mice. Experimental neurology 174, 193-200. Pavol, M.A., Meyers, C.A., Rexer, J.L., Valentine, A.D., Mattis, P.J., Talpaz, M., 1995. Pattern of neurobehavioral deficits associated with interferon alfa therapy for leukemia. Neurology 45, 947-950. Pedersen, L.H., Scheel-Kruger, J., Blackburn-Munro, G., 2007. Amygdala GABA-A receptor involvement in mediating sensory-discriminative and affective-motivational pain responses in a rat model of peripheral nerve injury. Pain 127, 17-26. Pellegrino, M.J., Parrish, D.C., Zigmond, R.E., Habecker, B.A., 2011. Cytokines inhibit norepinephrine transporter expression by decreasing Hand2. Molecular and cellular neurosciences 46, 671-680. Penzo, M.A., Robert, V., Li, B., 2014. Fear conditioning potentiates synaptic transmission onto long-range projection neurons in the lateral subdivision of central amygdala. The Journal of neuroscience : the official journal of the Society for Neuroscience 34, 2432-2437. Petrovich, G.D., Canteras, N.S., Swanson, L.W., 2001. Combinatorial amygdalar inputs to hippocampal domains and hypothalamic behavior systems. Brain research. Brain research reviews 38, 247-289. Plotkin, S.R., Banks, W.A., Kastin, A.J., 1996. Comparison of saturable transport and extracellular pathways in the passage of interleukin-1 alpha across the blood-brain barrier. J Neuroimmunol 67, 41-47. Qu, C., King, T., Okun, A., Lai, J., Fields, H.L., Porreca, F., 2011. Lesion of the rostral anterior cingulate cortex eliminates the aversiveness of spontaneous neuropathic pain following partial or complete axotomy. Pain 152, 1641-1648. Raison, C.L., Rutherford, R.E., Woolwine, B.J., et al., 2013. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: The role of baseline inflammatory biomarkers. JAMA Psychiatry 70, 3141. Rajasethupathy, P., Sankaran, S., Marshel, J.H., Kim, C.K., Ferenczi, E., Lee, S.Y., Berndt, A., Ramakrishnan, C., Jaffe, A., Lo, M., Liston, C., Deisseroth, K., 2015. Projections from neocortex mediate top-down control of memory retrieval. Nature 526, 653-659. Ren, W.-J., Liu, Y., Zhou, L.-J., Li, W., Zhong, Y., Pang, R.-P., Xin, W.-J., Wei, X.H., Wang, J., Zhu, H.-Q., Wu, C.-Y., Qin, Z.-H., Liu, G., Liu, X.-G., 2011. Peripheral Nerve Injury Leads to Working Memory Deficits and Dysfunction of the Hippocampus by Upregulation of TNF-alpha in Rodents. Neuropsychopharmacology, pp. 979-992.

44

Renno, W.M., 1998. Prolonged noxious stimulation increases periaqueductal gray NMDA mRNA expression: a hybridization study using two different rat models for nociception. Neurobiology 6, 333-357. Roozendaal, B., McReynolds, J.R., McGaugh, J.L., 2004. The basolateral amygdala interacts with the medial prefrontal cortex in regulating glucocorticoid effects on working memory impairment. The Journal of neuroscience : the official journal of the Society for Neuroscience 24, 1385-1392. Rothwell, N.J., Luheshi, G., Toulmond, S., 1996. Cytokines and their receptors in the central nervous system: physiology, pharmacology, and pathology. Pharmacology & therapeutics 69, 85-95. Sagheddu, C., Aroni, S., De Felice, M., Lecca, S., Luchicchi, A., Melis, M., Muntoni, A.L., Romano, R., Palazzo, E., Guida, F., Maione, S., Pistis, M., 2015. Enhanced serotonin and mesolimbic dopamine transmissions in a rat model of neuropathic pain. Neuropharmacology. Sahay, A., Hen, R., 2007. Adult hippocampal neurogenesis in depression. Nature neuroscience 10, 1110-1115. Salamone, J.D., Correa, M., Farrar, A., Mingote, S.M., 2007. Effort-related functions of nucleus accumbens dopamine and associated forebrain circuits. Psychopharmacology 191, 461-482. Samwel, H.J., Evers, A.W., Crul, B.J., Kraaimaat, F.W., 2006. The role of helplessness, fear of pain, and passive pain-coping in chronic pain patients. The Clinical journal of pain 22, 245-251. Sarkis, R., Saadé, N., Atweh, S., Jabbur, S., Al-Amin, H., 2011. Chronic dizocilpine or apomorphine and development of neuropathy in two rat models I: Behavioral effects and role of nucleus accumbens. Experimental neurology 228, 19-29. Schöbitz, B., de Kloet, E.R., Sutanto, W., Holsboer, F., 1993. Cellular Localization of Interleukin 6 mRNA and Interleukin 6 Receptor mRNA in Rat Brain. European Journal of Neuroscience 5, 1426-1435. Schwartz, N., Temkin, P., Jurado, S., Lim, B.K., Heifets, B.D., Polepalli, J.S., Malenka, R.C., 2014. Chronic pain. Decreased motivation during chronic pain requires long-term depression in the nucleus accumbens. Science 345, 535-542. Seltzer, Z., Dubner, R., Shir, Y., 1990. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 43, 205-218. Seminowicz, D.A., Laferriere, A.L., Millecamps, M., Yu, J.S., Coderre, T.J., Bushnell, M.C., 2009. MRI structural brain changes associated with sensory and emotional function in a rat model of long-term neuropathic pain. NeuroImage 47, 1007-1014. Shao, Q., Li, Y., Wang, Q., Zhao, J., 2015. IL-10 and IL-1beta Mediate NeuropathicPain Like Behavior in the Ventrolateral Orbital Cortex. Neurochemical research. Siren, A.L., McCarron, R., Wang, L., Garcia-Pinto, P., Ruetzler, C., Martin, D., Hallenbeck, J.M., 2001. Proinflammatory cytokine expression contributes to brain injury provoked by chronic monocyte activation. Molecular medicine 7, 219-229. Sloane, E., Langer, S., Jekich, B., Mahoney, J., Hughes, T., Frank, M., Seibert, W., Huberty, G., Coats, B., Harrison, J., Klinman, D., Poole, S., Maier, S., Johnson, K., Chavez, R., Watkins, L.R., Leinwand, L., Milligan, E., 2009. Immunological priming potentiates non-viral anti-inflammatory gene therapy treatment of neuropathic pain. Gene Ther 16, 1210-1222. Soderquist, R.G., Sloane, E.M., Loram, L.C., Harrison, J.A., Dengler, E.C., Johnson, S.M., Amer, L.D., Young, C.S., Lewis, M.T., Poole, S., Frank, M.G., Watkins, L.R., Milligan, E.D., Mahoney, M.J., 2010. Release of plasmid DNA-encoding IL-10 from

45

PLGA microparticles facilitates long-term reversal of neuropathic pain following a single intrathecal administration. Pharm Res 27, 841-854. Spengler, R.N., Sud, R., Knight, P.R., Ignatowski, T.A., 2007. Antinociception mediated by alpha(2)-adrenergic activation involves increasing tumor necrosis factor alpha (TNFalpha) expression and restoring TNFalpha and alpha(2)-adrenergic inhibition of norepinephrine release. Neuropharmacology 52, 576-589. Stefanik, M.T., Kalivas, P.W., 2013. Optogenetic dissection of basolateral amygdala projections during cue-induced reinstatement of cocaine seeking. Frontiers in behavioral neuroscience 7, 213. Stellwagen, D., Beattie, E.C., Seo, J.Y., Malenka, R.C., 2005. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. The Journal of neuroscience : the official journal of the Society for Neuroscience 25, 3219-3228. Stuart, M.J., Baune, B.T., 2014. Chemokines and chemokine receptors in mood disorders, schizophrenia, and cognitive impairment: A systematic review of biomarker studies. Neuroscience & Biobehavioral Reviews 42, 93-115. Sud, R., Spengler, R.N., Nader, N.D., Ignatowski, T.A., 2008. Antinociception occurs with a reversal in alpha 2-adrenoceptor regulation of TNF production by peripheral monocytes/macrophages from pro- to anti-inflammatory. European journal of pharmacology 588, 217-231. Sugam, J.A., Day, J.J., Wightman, R.M., Carelli, R.M., 2012. Phasic nucleus accumbens dopamine encodes risk-based decision-making behavior. Biological psychiatry 71, 199-205. Swanson, L.W., 2000. Cerebral hemisphere regulation of motivated behavior. Brain research 886, 113-164. Tajerian, M., Alvarado, S., Millecamps, M., Vachon, P., Crosby, C., Bushnell, M.C., Szyf, M., Stone, L.S., 2013. Peripheral nerve injury is associated with chronic, reversible changes in global DNA methylation in the mouse prefrontal cortex. PloS one 8, e55259. Takao, T., Tracey, D.E., Mitchell, W.M., De Souza, E.B., 1990. Interleukin-1 receptors in mouse brain: characterization and neuronal localization. Endocrinology 127, 3070-3078. Takeda, K., Muramatsu, M., Chikuma, T., Kato, T., 2009. Effect of memantine on the levels of neuropeptides and microglial cells in the brain regions of rats with neuropathic pain. Journal of molecular neuroscience : MN 39, 380-390. Tanabe, M., Takasu, K., Yamaguchi, S., Kodama, D., Ono, H., 2008. Glycine transporter inhibitors as a potential therapeutic strategy for chronic pain with memory impairment. Anesthesiology 108, 929-937. Tancredi, V., D'Arcangelo, G., Grassi, F., Tarroni, P., Palmieri, G., Santoni, A., Eusebi, F., 1992. Tumor necrosis factor alters synaptic transmission in rat hippocampal slices. Neuroscience letters 146, 176-178. Taylor, A.M., Castonguay, A., Taylor, A.J., Murphy, N.P., Ghogha, A., Cook, C., Xue, L., Olmstead, M.C., De Koninck, Y., Evans, C.J., Cahill, C.M., 2015. Microglia disrupt mesolimbic reward circuitry in chronic pain. The Journal of neuroscience : the official journal of the Society for Neuroscience 35, 8442-8450. Taylor, A.M., Murphy, N.P., Evans, C.J., Cahill, C.M., 2014. Correlation between ventral striatal catecholamine content and nociceptive thresholds in neuropathic mice. The journal of pain : official journal of the American Pain Society 15, 878-885. Terada, M., Kuzumaki, N., Hareyama, N., Imai, S., Niikura, K., Narita, M., Yamazaki, M., Suzuki, T., Narita, M., 2008. Suppression of enriched environment-

46

induced neurogenesis in a rodent model of neuropathic pain. Neuroscience letters 440, 314-318. Terashima, T., Shirakawa, K., Maekawa, M., Furukawa, N., Yamaguchi, S., Hori, Y., 2012. Differential expression of NMDA receptors in serotonergic and/or GABAergic neurons in the midbrain periaqueductal gray of the mouse. Neuroscience letters 528, 55-60. Tilleux, S., Berger, J., Hermans, E., 2007. Induction of astrogliosis by activated microglia is associated with a down-regulation of metabotropic glutamate receptor 5. J Neuroimmunol 189, 23-30. Treede, R.D., Jensen, T.S., Campbell, J.N., Cruccu, G., Dostrovsky, J.O., Griffin, J.W., Hansson, P., Hughes, R., Nurmikko, T., Serra, J., 2008. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 70, 1630-1635. Tsai, Y.J., Huang, C.T., Lin, S.C., Yeh, J.H., 2012. Effects of regional and wholebody hypothermic treatment before and after median nerve injury on neuropathic pain and glial activation in rat cuneate nucleus. Anesthesiology 116, 415-431. Uceyler, N., Rogausch, J.P., Toyka, K.V., Sommer, C., 2007. Differential expression of cytokines in painful and painless neuropathies. Neurology 69, 42-49. Uceyler, N., Tscharke, A., Sommer, C., 2008. Early cytokine gene expression in mouse CNS after peripheral nerve lesion. Neuroscience letters 436, 259-264. Ulrich-Lai, Y.M., Xie, W., Meij, J.T., Dolgas, C.M., Yu, L., Herman, J.P., 2006. Limbic and HPA axis function in an animal model of chronic neuropathic pain. Physiology & behavior 88, 67-76. Utsuyama, M., Hirokawa, K., 2002. Differential expression of various cytokine receptors in the brain after stimulation with LPS in young and old mice. Experimental gerontology 37, 411-420. Van Dam, A.M., De Vries, H.E., Kuiper, J., Zijlstra, F.J., De Boer, A.G., Tilders, F.J., Berkenbosch, F., 1996. Interleukin-1 receptors on rat brain endothelial cells: a role in neuroimmune interaction? FASEB journal : official publication of the Federation of American Societies for Experimental Biology 10, 351-356. van der Meer, P., Ulrich, A.M., Gonźalez-Scarano, F., Lavi, E., 2000. Immunohistochemical Analysis of CCR2, CCR3, CCR5, and CXCR4 in the Human Brain: Potential Mechanisms for HIV Dementia. Experimental and Molecular Pathology 69, 192-201. Vitkovic, L., Bockaert, J., Jacque, C., 2000. "Inflammatory" cytokines: neuromodulators in normal brain? Journal of neurochemistry 74, 457-471. Viviani, B., Bartesaghi, S., Gardoni, F., Vezzani, A., Behrens, M.M., Bartfai, T., Binaglia, M., Corsini, E., Di Luca, M., Galli, C.L., Marinovich, M., 2003. Interleukin1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. The Journal of neuroscience : the official journal of the Society for Neuroscience 23, 8692-8700. Walker, A.K., Budac, D.P., Bisulco, S., Lee, A.W., Smith, R.A., Beenders, B., Kelley, K.W., Dantzer, R., 2013. NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 38, 1609-1616. Walker, A.K., Kavelaars, A., Heijnen, C.J., Dantzer, R., 2014. Neuroinflammation and comorbidity of pain and depression. Pharmacological reviews 66, 80-101. Walton, M.E., Bannerman, D.M., Alterescu, K., Rushworth, M.F., 2003. Functional specialization within medial frontal cortex of the anterior cingulate for evaluating

47

effort-related decisions. The Journal of neuroscience : the official journal of the Society for Neuroscience 23, 6475-6479. Wang, J., Liu, Y., Zhou, L.J., Wu, Y., Li, F., Shen, K.F., Pang, R.P., Wei, X.H., Li, Y.Y., Liu, X.G., 2013a. Magnesium L-threonate prevents and restores memory deficits associated with neuropathic pain by inhibition of TNF-alpha. Pain physician 16, E563-575. Wang, J., Yu, J., Ding, C.P., Han, S.P., Zeng, X.Y., Wang, J.Y., 2015a. Transforming growth factor-beta in the red nucleus plays antinociceptive effect under physiological and pathological pain conditions. Neuroscience 291, 37-45. Wang, X.M., Liu, W.J., Zhang, R., Zhou, Y.K., 2013b. Effects of exposure to lowlevel lead on spatial learning and memory and the expression of mGluR1, NMDA receptor in different developmental stages of rats. Toxicology and industrial health 29, 686-696. Wang, X.Q., Zhong, X.L., Li, Z.B., Wang, H.T., Zhang, J., Li, F., Zhang, J.Y., Dai, R.P., Xin-Fu, Z., Li, C.Q., Li, Z.Y., Bi, F.F., 2015b. Differential roles of hippocampal glutamatergic receptors in neuropathic anxiety-like behavior after partial sciatic nerve ligation in rats. BMC neuroscience 16, 14. Wang, Z., Wang, J., Li, X., Yuan, Y., Fan, G., 2008. Interleukin-1 beta of Red nucleus involved in the development of allodynia in spared nerve injury rats. Experimental brain research 188, 379-384. Wang, Z.H., Zeng, X.Y., Han, S.P., Fan, G.X., Wang, J.Y., 2012. Interleukin-10 of red nucleus plays anti-allodynia effect in neuropathic pain rats with spared nerve injury. Neurochemical research 37, 1811-1819. Wei, F., Guo, W., Zou, S., Ren, K., Dubner, R., 2008. Supraspinal glial-neuronal interactions contribute to descending pain facilitation. The Journal of neuroscience : the official journal of the Society for Neuroscience 28, 10482-10495. Wu, H.Q., Rassoulpour, A., Schwarcz, R., 2007. Kynurenic acid leads, dopamine follows: a new case of volume transmission in the brain? Journal of neural transmission 114, 33-41. Wu, Y., Na, X., Zang, Y., Cui, Y., Xin, W., Pang, R., Zhou, L., Wei, X., Li, Y., Liu, X., 2014. Upregulation of tumor necrosis factor-alpha in nucleus accumbens attenuates morphine-induced rewarding in a neuropathic pain model. Biochemical and biophysical research communications 449, 502-507. Yamashita, A., Hamada, A., Suhara, Y., Kawabe, R., Yanase, M., Kuzumaki, N., Narita, M., Matsui, R., Okano, H., Narita, M., 2014. Astrocytic activation in the anterior cingulate cortex is critical for sleep disorder under neuropathic pain. Synapse 68, 235-247. Yan, H.Q., Banos, M.A., Herregodts, P., Hooghe, R., Hooghe-Peters, E.L., 1992. Expression of interleukin (IL)-1β, IL-6 and their respective receptors in the normal rat brain and after injury. European Journal of Immunology 22, 2963-2971. Yang, L., Kress, B.T., Weber, H.J., Thiyagarajan, M., Wang, B., Deane, R., Benveniste, H., Iliff, J.J., Nedergaard, M., 2013. Evaluating glymphatic pathway function utilizing clinically relevant intrathecal infusion of CSF tracer. Journal of translational medicine 11, 107. Yang, S., Liu, Z.W., Wen, L., Qiao, H.F., Zhou, W.X., Zhang, Y.X., 2005. Interleukin-1beta enhances NMDA receptor-mediated current but inhibits excitatory synaptic transmission. Brain research 1034, 172-179. Zeng, X.Y., Zhang, Q., Wang, J., Yu, J., Han, S.P., Wang, J.Y., 2014. Distinct role of tumor necrosis factor receptor subtypes 1 and 2 in the red nucleus in the development of neuropathic pain. Neuroscience letters 569, 43-48.

48

Zhang, F., Vadakkan, K.I., Kim, S.S., Wu, L.J., Shang, Y., Zhuo, M., 2008. Selective activation of microglia in spinal cord but not higher cortical regions following nerve injury in adult mouse. Molecular pain 4, 15. Zhou, W., Dantzer, R., Budac, D.P., Walker, A.K., Mao-Ying, Q.-L., Lee, A.W., Heijnen, C.J., Kavelaars, A., 2015. Peripheral indoleamine 2,3-dioxygenase 1 is required for comorbid depression-like behavior but does not contribute to neuropathic pain in mice. Brain, behavior, and immunity 46, 147-153. Zhu, C.B., Lindler, K.M., Owens, A.W., Daws, L.C., Blakely, R.D., Hewlett, W.A., 2010. Interleukin-1 receptor activation by systemic lipopolysaccharide induces behavioral despair linked to MAPK regulation of CNS serotonin transporters. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 35, 2510-2520.

49

Figure legends Figure 1. A schematic overview showing three proposed routes of immune-to-brain signalling which can lead to brain neuroinflammation following a peripheral immune challenge. (a) Neural transmission; the most common example is via the vagus nerve, where IL-1β activates vagal afferents, altering the firing pattern of neurons in the nucleus tractus solitarius (NTS) and its projection targets the amygdala and hypothalamus, leading to the induction of cytokine release by neighbouring glial cells (Brady et al., 1994; Goehler et al., 1999; Hansen et al., 2001). (b) Humoral transmission; pro-inflammatory cytokines enter systemic circulation and are either actively transported across the BBB, pass through into CVOs or act on astrocytes and endothelial cells within the BBB to induce their production in the brain, either directly or indirectly via the release of nitric oxide (NO) and PGs (Banks et al., 1994; Banks et al., 1991; Konsman et al., 1999; Konsman et al., 2002; Pan and Kastin, 2002; Plotkin et al., 1996; Van Dam et al., 1996). (c) Molecular transmission; systemic cytokines, such as TNF, trigger the release of MCP-1 from microglia within the brain that facilitates monocyte adhesion and rolling along the endothelial cells, thus allowing monocyte infiltration across the BBB which results in the propagation of neuroinflammation (D'Mello et al., 2009; D'Mello et al., 2013; Mitchell et al., 2009).

Figure 2. A schematic overview of five possible mechanisms by which either peripheral inflammation or supraspinal neuroinflammatory mediators may underlie changes in affective behaviour. (i) Altered glutamatergic and GABAergic activity, resulting in a reduction in LTP and excitotoxicity resulting in neuronal and glial damage (Beattie et al., 2002; Bezzi et al., 2001; Furukawa and Mattson, 1998; Ida et al., 2008; Ren et al., 2011; Stellwagen et al., 2005; Tilleux et al., 2007; Wang et al.,

50

2013b; Yang et al., 2005). (ii) An increase in production of the enzyme, indoleamine 2,3-dioxygenase (IDO) that facilitates the conversion of tryptophan to kynurenine (KYN). In microglia KYN can be converted to the neurotoxic metabolite QUIN, an agonist of the NMDA receptor (Guillemin et al., 2001). Increased expression of KYN and QUIN has been linked to depression in humans, as well as depressive-like behaviour in neuropathic and inflammatory chronic pain models (Kim et al., 2012; O'Connor et al., 2009; Walker et al., 2013; Zhou et al., 2015). (iii) Suppression of growth factors, particularly BDNF, leading to a reduction in neurogenesis (Goshen et al., 2008; Goshen et al., 2007). (iv) Changes in monoamine turnover (Covey et al., 2000; Moron et al., 2003; Pellegrino et al., 2011; Wu et al., 2007; Zhu et al., 2010). (v) Reduced glucocorticoid release as well as a decrease in GR expression and function (Besedovsky and del Rey, 1996; Pace et al., 2007).

51

52

53

Table 1. Summary of previous studies on the time course of neuroinflammation following peripheral nerve injury. Brain region Time Nerve Species/Strain/ Neuroinflammation period injury Sex model Hippocampus <24 SNI Rat Increased TNF protein hours SpragueDawley Male CCI Mice Decreased TNF, IL-1β, and IL-4 mRNA C57BL/6J Female 3-4 days SNI Rat Increased TNF protein SpragueDawley Male 1-2 CCI and Rat Increased bilateral IL-1β, and increased weeks SNI Spraguecontralateral IL-6 protein after both CCI and Dawley SNI Female CCI Rat Increased TNF protein, which disappears at day Sprague14 Dawley Male CCI and Rat Increased contralateral IL-1β and IL-6 mRNA SNI Spragueafter SNI but not CCI in Wistar-Kyoto Dawley/ Increased contralateral IL-1β after both SNI and Wistar-Kyoto CCI but no change in IL-6 mRNA in SpragueMale Dawley CCI Rat Increased contralateral TNF protein SpragueDawley Male CCI Rat Increased TNF protein SpragueDawley Male Sciatic Mouse Increased TNF protein nerve Swiss crush Male SNI Rat Increased TNF protein SpragueDawley Male CCI Rat Increased TNF mRNA SpragueDawley Male CCI Rat Increased contralateral TNF protein SpragueDawley Male SNI Rat Increased TNF protein SpragueDawley Male Common Mouse No change in microglia activation peroneal Cx3cr1GFP/+

54

Reference

(Ren et al., 2011)

(Uceyler et al., 2008) (Ren et al., 2011)

(Al-Amin et al., 2011)

(Covey et al., 2000)

(del Rey et al., 2011)

(Gerard et al., 2015)

(Ignatowski et al., 1999)

(Nascimento et al., 2015) (Ren et al., 2011)

(Spengler et al., 2007)

(Sud et al., 2008)

(Wang et al., 2013)

(Zhang et al., 2008)

3+ weeks

nerve ligation SNL

CCI and SNI

SNI

SNI

PFC

<24 hours

CCI

1-2 weeks

CCI and SNI

CCI and SNI SNI

SNI

SNL

SNI

PSNL

Sciatic nerve crush SNI

SNI

SNI

Not stated Rat SpragueDawley Male Rat SpragueDawley/ Wistar Kyoto Male

Rat SpragueDawley Male Rat SpragueDawley Male Mice C57BL/6J Female Rat SpragueDawley Female Rat Wistar Kyoto Male Mouse C57BL/6N Male Mouse C57BL/6N Male Rat SpragueDawley Male Rat SpragueDawley Male Mouse C57BL/6J Male Mouse Swiss Male Mouse C57BL/6 Male Mouse C57BL/6 Male Rat Sprague-

Increased astrocyte expression when combined with MD

(Burke et al., 2013b)

Increased contralateral IL-1β but disappearance of increased IL-6 mRNA after SNI in Wistar Kyoto Increased contralateral IL-1β after SNI but disappearance of increased IL-1β mRNA after CCI, and increased IL-1ra mRNA after both SNI and CCI in Sprague-Dawley Increased TNF protein

(del Rey et al., 2011)

(Ren et al., 2011)

Increased TNF protein

(Wang et al., 2013)

Decreased TNF, but not IL-1β, mRNA

(Uceyler et al., 2008)

Increased bilateral IL-1β and contralateral IL-6 protein after both SNI and CCI (ACC)

(Al-Amin et al., 2011)

Increased contralateral IL-1β mRNA after SNI but not CCI

(Apkarian et al., 2006)

Increased IL-1β protein (OFC)

(Fuccio et al., 2009)

Increased IL-1R1 but not IL-1β protein (PL-IL)

(Giordano et al., 2012)

Increased contralateral IL-1β, IL-6 and TNF but not IL-10 protein

(Liu et al., 2007)

No change in astrocyte or microglia expression (mPFC)

(Marcello et al., 2013)

Increased astrocyte expression and activation, but no change in mRNA (ACC)

(Narita et al., 2011)

Increased TNF protein

(Nascimento et al., 2015)

Increased IL-1β but not IL-6, TNF and astrocyte mRNA when also socially isolated

(Norman et al., 2010a)

Increased IL-1β but not IL-6, TNF and astrocyte mRNA

(Norman et al., 2010b)

Increased contralateral IL-1β and IL-10 protein (VLO)

(Shao et al., 2015)

55

PSNL

3+ weeks

Common peroneal nerve ligation CCI and SNI PSNL

Striatum/ NAcc

1-2 weeks

CCI and SNI

CCI and SNI SNI

SNI

VTA

3+ weeks

CCI and SNI

1-2 weeks

PNI

SNI

Amygdala

1-2 weeks

3+ weeks

Hypothalamus

SNI

Common peroneal nerve ligation SNL

<24 hours

CCI

3-4 days

PSNL

1-2

CCI

Dawley Male Mouse C57BL/6J Male Mouse Cx3cr1GFP/+ Not stated

Increased astrocyte expression and activation after a thermal noxious stimulus (ACC)

(Yamashita et al., 2014)

No change in microglia activation (ACC and PFC)

(Zhang et al., 2008)

Rat Wistar Kyoto Male Mouse C57BL/6J Male Rat SpragueDawley Female Rat Wistar Kyoto Male Rat SpragueDawley Male Rat SpragueDawley Male Rat Wistar Kyoto Male Mouse C57BL/6J Male Rat SpragueDawley Male Rat SpragueDawley Male Mouse Cx3cr1GFP/+ Not stated

Disappearance of increased contralateral IL-1β mRNA after SNI, no change after CCI

(Apkarian et al., 2006)

Increased astrocyte expression (ACC)

(Narita et al., 2006)

Increased bilateral IL-1β after both CCI and SNI, and decreased contralateral IL-6 protein after CCI (Striatum)

(Al-Amin et al., 2011)

Decreased contralateral IL-1β mRNA after both CCI and SNI (Striatum/thalamus)

(Apkarian et al., 2006)

No change in astrocyte or microglia expression (NAcc)

(Marcello et al., 2013)

Increased bilateral TNF protein (NAcc)

(Wu et al., 2014)

Increased contralateral IL-1β after SNI and disappearance of decreased contralateral IL-1β mRNA after CCI (Striatum/thalamus) Increased microglial activation

(Apkarian et al., 2006)

Rat SpragueDawley Male Mice C57BL/6J Female Rat Wistar Male Rat

(Taylor et al., 2015)

No change in bilateral TNF protein

(Wu et al., 2014)

Increased astrocyte expression, but no change in microglia expression

(Marcello et al., 2013)

No change in microglia activation

(Zhang et al., 2008)

Increased IL-10 and decreased IL-6, but no change in IL-1β, TNF, astrocyte or microglia mRNA

(Burke et al., 2013a)

Decreased TNF and IL-4, but no change IL-1β, mRNA

(Uceyler et al., 2008)

Increased microglial expression

(Takeda et al., 2009)

No change in IL-1β or TNF mRNA

(Kilburn-

56

weeks

PAG

3-4 days

CCI

PSNL

1-2 weeks

CCI

SNI

CCI

SNI

Common peroneal nerve ligation

SpragueDawley Male Rat Wistar Male Rat Wistar Male Rat Wistar Male Rat SpragueDawley Male Rat SpragueDawley Male Mouse C57BL/6 Male Mouse Cx3cr1GFP/+ Not stated

Watt et al., 2014) Increased astrocyte, microglia, TNF, IL-1β and IL-6, but no change in IL-10 protein

(Chu et al., 2012)

Increased microglial expression

(Takeda et al., 2009)

Increased astrocyte, microglia, TNF, IL-1β and IL-6, but no change in IL-10 protein

(Chu et al., 2012)

No change in astrocyte or microglia expression

(Marcello et al., 2013)

Increased astrocyte expression (lPAG and vlPAG)

(Mor et al., 2010)

Increased astrocyte, but no change in IL-1β, IL6 and TNF mRNA

(Norman et al., 2010b)

No change in microglia activation

(Zhang et al., 2008)

57

Table 2. Summary of previous studies on the effects of neuroinflammation on affective behaviour post-injury. Brain Region Nerve Species/ Affective Key findings Reference injury Strain/ behavioural model Sex test Hippocampus SNI Mouse Sucrose Decreased sucrose preference (Dellarole et C57BL/6 preference causally related to an increase al., 2014) Not stated in TNF and TNFR1 signalling Hippocampus

SNI

Rat SpragueDawley Male Rat SpragueDawley Male Mouse Swiss Male

Radial maze Novel object recognition Novel object recognition

Impaired short-term spatial and novelty recognition memory causally related to an increase in TNF Impaired short-term novelty recognition memory causally related to an increase in TNF

Hippocampus

SNI

Hippocampus and PFC

Crush injury

mPFC (ACC)

FST TST

Increased immobility causally related to an increase in TNF

(Nascimento et al., 2015)

PSNL

Mouse C57BL/6J Male

Light-dark box EPM

(Narita et al., 2006)

mPFC (ACC)

PSNL

Mouse C57BL/6J Male

EEG/EMG

PFC

SNI

Mouse C57BL/6 Male

FST

Decreased time spent in light compartment and in open arms causally related to an increase in astrocytes Increased wakefulness and decreased NREM related to an increase in astrocyte expression and activation Increased immobility related to an increase in IL-1β and IL-1R1 signalling

NAcc

SNI

CPP

PNI

Prevention of CPP to low dose morphine causally related to an increase in TNF and TNFR1 signalling Prevention of CPP to low dose synthetic opioid causally related to an increase in microglial activation

(Wu et al., 2014)

VTA

Amygdala

SNL

Rat SpragueDawley Male Rat/Mouse SpragueDawley/ C57BL/6J Male/Male Rat SpragueDawley Male

(Burke et al., 2013a)

PAG

CCI

Rat SpragueDawley Male

Residentintruder paradigm

Decreased time spent in the inner zone related to an increase in IL-10 and decrease in IL-6 gene expression at a later time point Decreased dominance behaviour related to an increase in astrocyte expression within the vlPAG in a sub-group of rats

CPP

Open field

58

(Ren et al., 2011)

(Wang et al., 2013)

(Narita et al., 2011)

(Norman et al., 2010b)

(Taylor et al., 2015)

(Mor et al., 2010)

Highlights • Peripheral inflammation results in affective disturbances in neuropathic pain • Nerve injury induces increases in brain cytokine expression and glial cell activity • Inflammation in hippocampal-prefrontal circuits causes affective disturbances • Anti-inflammatories may prevent affective disturbances in neuropathic pain

59