Immune and inflammatory mechanism in neuropathic pain

Embed Size (px)

Citation preview

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    1/25

    Review

    Immune and inflammatory mechanisms in neuropathic pain

    Gila Moalem, David J. Tracey

    School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia

    A R T I C L E I N F O A B S T R A C T

    Article history:

    Accepted 17 November 2005

    Available online 4 January 2006

    Tissue damage, inflammation or injury of the nervous system may result in chronic

    neuropathic pain characterised by increased sensitivity to painful stimuli (hyperalgesia), the

    perception of innocuous stimuli as painful (allodynia) and spontaneous pain. Neuropathic

    pain has been described in about 1% of the US population, is often severely debilitating and

    largely resistant to treatment. Animal models of peripheral neuropathic pain are now

    available in which the mechanisms underlying hyperalgesia and allodynia due to nerve

    injury or nerve inflammation can be analysed. Recently, it has become clear that

    inflammatory and immune mechanisms both in the periphery and the central nervous

    system play an important role in neuropathic pain. Infiltrationof inflammatory cells, as well

    as activation of resident immune cells in response to nervous system damage, leads to

    subsequent production and secretion of various inflammatory mediators. These mediators

    promote neuroimmune activation and can sensitise primary afferent neurones and

    contribute to pain hypersensitivity. Inflammatory cells such as mast cells, neutrophils,macrophages and T lymphocytes have all been implicated, as have immune-like glial cells

    such as microglia and astrocytes. In addition, the immune response plays an important role

    in demyelinating neuropathies such as multiple sclerosis (MS), in which pain is a common

    symptom, and an animal model of MS-related pain has recently been demonstrated. Here,

    we will briefly review some of the milestones in research that have led to an increased

    awareness of the contribution of immune and inflammatory systems to neuropathic pain

    and then review in more detail the role of immune cells and inflammatory mediators.

    2005 Elsevier B.V. All rights reserved.

    Keywords:

    Neuropathic pain

    Nerve injury

    Immunity

    Inflammation

    Abbreviations:

    5HT, 5-hydroxytryptamine,

    serotonin8R, 15S-diHETE, (8R, 15S)-

    dihydroxyeicosa-(5E-9,11,13Z)-

    tetraenoic acid

    ADP, adenosine diphosphate

    AMP, adenosine monophosphate

    ATP, adenosine triphosphate

    BDNF, brain-derived neurotrophic

    factor

    CCR2, chemokine (CC motif)

    receptor 2

    CGRP, calcitonin-gene-related

    peptide

    COX-1, cyclooxygenase-1COX-2, cyclooxygenase-2

    CX3CR1, G-protein-coupled

    chemokine receptor for fractalkine

    DRG, dorsal root ganglion

    GABA, gamma aminobutyric acid

    GBS, GuillainBarr syndrome

    GDNF, glial-cell-line-derived

    neurotrophic factor

    B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

    Corresponding author. Fax: +61 2 9385 8016.E-mail address: [email protected] (D.J. Tracey).

    0165-0173/$ see front matter 2005 Elsevier B.V. All rights reserved.doi:10.1016/j.brainresrev.2005.11.004

    a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m

    w w w . e l s e v i e r . c o m / l o c a t e / b r a i n r e s r e v

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    2/25

    IL-1, interleukin-1

    L-NAME, NG-nitro-L-arginine methyl

    ester

    LTB4, leukotriene B4MCP-1, monocyte chemoattractant

    protein-1

    MHC, major histocompatibility

    complex

    MIP-1, macrophage inflammatory

    protein-1

    MRP-14, cytosolic-calcium-binding

    protein of 14 kDa

    MS, multiple sclerosis

    NGF, nerve growth factor

    NO, nitric oxide

    NOS, nitric oxide synthase

    NSAID, non-steroidal

    anti-inflammatory drug

    NT-3, Neurotrophin-3

    NT-4/5, Neurotrophin-4/5

    P2X, ATP-gated ion channels

    P2Y, G-protein-coupled ion channels

    activated by purine/pyrimidine

    nucleotides

    PAR-2, protease-activated receptor-2

    PGE2, Prostaglandin E2PGI2, Prostaglandin I2TCA, tricyclic antidepressant

    TEMPOL, 4-hydroxy-2,2,6,6-

    tetramethylpiperidine-1-oxyl

    Th1 cells, T helper 1 cells

    Th2 cells, T helper 2 cells

    TLR4, Toll-like receptor 4

    TNF, tumour necrosis factor

    TrkA, tyrosine kinase receptor A

    TrkB, tyrosine kinase receptor B

    TrkC, tyrosine kinase receptor C

    TRPV1, transient receptor potential

    (receptor) vanilloid 1

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242

    1.1. Chemical environment of sensory neurones contributes to neuropathic pain . . . . . . . . . . . . . . . . . 2421.2. Inflammatory models of neuropathic pain. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243

    1.3. Neuropathic pain in autoimmune diseases of the nervous system . . . . . . . . . . . . . . . . . . . . . . . 243

    1.4. Direct and indirect actions of mediators released by immune cells . . . . . . . . . . . . . . . . . . . . . . . 243

    2. Immune cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 244

    2.1. Mast cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 244

    2.2. Neutrophils . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245

    2.3. Macrophages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247

    2.4. Lymphocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247

    2.5. Glia and Schwann cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248

    3. Mediators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249

    3.1. Bradykinin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249

    3.2. ATP and adenosine. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249

    3.3. Serotonin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250

    241B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    3/25

    3.4. Eicosanoids. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250

    3.5. Cytokines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251

    3.5.1. Interleukin-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 52

    3.5.2. Interleukin-6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 52

    3.5.3. Tumour necrosis factor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252

    3.5.4. Interleukin-10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 53

    3.6. Neurotrophins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253

    3.7. Nitric oxide and reactive oxygen species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2544. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255

    References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255

    1. Introduction

    Neuropathic pain is caused by lesion or inflammation of

    the nervous system and is relatively common, with an

    incidence estimated at 0.6% to 1.5% in the US population

    (Warfield and Fausett, 2002). It is often severely debilitating

    and largely resistant to treatment (Harden and Cohen,

    2003) mainly because the underlying mechanisms are still

    poorly understood. Symptoms of neuropathic pain may

    include allodynia (pain resulting from a stimulus that is

    normally non-painful), hyperalgesia (an excessive response

    to painful stimuli) and spontaneous pain. The study of

    neuropathic pain can be traced back to Weir Mitchell and

    his classic work on nerve injuries from the American Civil

    War (Mitchell, 1872). Since that time, a great deal has been

    written on neuropathic pain and its possible causes, but it

    was only with the development of animals models of pain

    due to nerve injury that some real progress was made in

    understanding some of the mechanisms involved. For

    recent reviews of these mechanisms, see Julius and

    Basbaum (2001) and Scholz and Woolf (2002). The first

    widely used animal model of neuropathic pain was chronic

    constriction injury of the rat sciatic nerve (Bennett and Xie,

    1988), in which the sciatic nerve is encircled by four

    ligatures of chromic gut. This leads to the cardinal

    symptoms of neuropathic painhyperalgesia, allodynia

    and apparent spontaneous pain. Other widely used animal

    models include partial ligation of the sciatic nerve (Seltzer

    et al., 1990) and section of one or more of the spinal

    nerves which contribute to the sciatic (Kim and Chung,

    1992). Research on these animal models of peripheral

    neuropathic pain (the pain syndrome resulting from

    lesions of the peripheral nervous system) has made it

    clear that a number of mechanisms are involved, including

    ectopic excitability of sensory neurones, altered gene

    expression of sensory neurones and sensitisation of

    neurones in the dorsal horn of the spinal cord (Scholz

    and Woolf, 2002; Woolf, 2004). However, there is increasing

    evidence that inflammatory and immune mechanisms also

    play a role, and we will begin by looking at a brief history

    of how this evidence has accumulated. We will then look

    in more detail at the evidence for involvement of immune

    cells and inflammatory mediators in neuropathic pain.

    However, the review does not provide exhaustive coverage

    of all immune and inflammatory mechanisms that con-

    tribute to pain following nerve injury. For readers who

    would like to know more about these and other aspects of

    neuropathic pain, several recent reviews are available,

    dealing with immune and glial mechanisms (Watkins and

    Maier, 2002, 2005; DeLeo et al., 2004; McMahon et al., 2005;

    Tsuda et al., 2005), inflammatory aspects (Woolf and

    Costigan, 1999; DeLeo and Yezierski, 2001; DeLeo et al.,

    2004; Sommer and Kress, 2004) and cytokines (Sommer andKress, 2004).

    1.1. Chemical environment of sensory neurones

    contributes to neuropathic pain

    At the time that the first animal models of pain due to

    nerve injury were developed, it was widely believed that

    injury or loss of myelinated and unmyelinated sensory

    axons in the sciatic nerve was the key factor in producing

    symptoms of neuropathic pain. However, some studies in

    the early 1990s suggested that other factors might also be

    involved. For example, Maves showed that chromic gut

    alone placed next to the sciatic nerve induced thermal

    hyperalgesia without any loss of myelinated axons (Maves

    et al., 1993). Of course, it had long been known that C-

    polymodal nociceptors are sensitised or activated by

    chemical stimuli, including inflammatory mediators (Wood

    and Docherty, 1997), but these effects were thought of as

    being responsible for inflammatory pain, and little atten-

    tion was given to the idea that immune or inflammatory

    cells or their mediators might play a role in neuropathic

    pain until the advent of useful animal models. It was also

    known that nerve injury resulted in activation of mast cells

    (Olsson, 1967) and recruitment of neutrophils and macro-

    phages (Perry et al., 1987). However, it was not until 1993

    that acute changes in the endoneurial microenvironment

    were explicitly linked with the initial development of

    hyperalgesia (Frisen et al., 1993; Sommer et al., 1993).

    Further work confirmed suggestions that the factors

    responsible for neuropathic hyperalgesia included Wallerian

    degeneration and macrophage activation (Sommer et al.,

    1995). At around the same time, Clatworthy followed up the

    observations of Maves et al. and showed that suppression

    of the inflammatory response in the injured sciatic nerve

    blocked the development of hyperalgesia, while enhancing

    the inflammatory response augmented the hyperalgesia

    (Clatworthy et al., 1995). Evidence on the involvement of

    immune cells and inflammatory mediators in hyperalgesia

    was reviewed at this time, and it was suggested that these

    242 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    4/25

    mediators (and cytokines in particular) were a probable link

    between the activation or recruitment of immune cells to

    the injured nerve and the development of hyperalgesia and

    neuropathic pain (Tracey and Walker, 1995; Watkins et al.,

    1995). Glial cells supporting neurones in the spinal cord

    (Meller et al., 1994) and Schwann cells supporting axons in

    the peripheral nerve (Constable et al., 1994; Wagner and

    Myers, 1996a) were added to the list of cell types whichmight contribute to neuropathic pain. In fact, glial cells and

    Schwann cells may also play an active role in the immune

    process by releasing mediators, including cytokines, and

    acting as antigen-presenting cells following nerve injury

    (Argall et al., 1992; Bergsteinsdottir et al., 1992; Constable et

    al., 1994). These ideas were not widely accepted at the

    time, but, gradually, evidence accumulated to give them

    further support. For example, tumour necrosis factor (TNF)

    and interleukins 1 and 6 (IL-1, IL-6) had already been

    shown to induce acute or short-term hyperalgesia (Ferreira

    et al., 1988; Cunha et al., 1992) but were now implicated

    directly in neuropathic pain, involving chronic hyperalgesia

    and allodynia (Wagner and Myers, 1996b; DeLeo et al., 1997;

    Arruda et al., 1998).

    Another example is the contribution of nerve growth

    factor (NGF), which shares many attributes with typical

    cytokines (Bonini et al., 2003). NGF is best known for its

    role in protecting some neuronal types from cell death

    during development, but Levine and his colleagues showed

    that the N-terminal octapeptide of NGF elicited hyperalge-

    sia in the rat when applied to injured tissue (Taiwo et al.,

    1991). These experiments were provoked by apparent

    similarities between the NGF fragment and bradykininan

    inflammatory mediator known to activate or sensitise

    nociceptors and to elicit hyperalgesia (Dray and Perkins,

    1993; Dray, 1997). By this stage, it was known that the level

    of NGF rises substantially in inflamed tissue and in injured

    nerve (Heumann et al., 1987; Weskamp and Otten, 1987),

    and a causal relationship between tissue levels of NGF and

    inflammatory hyperalgesia was confirmed by showing that

    anti-NGF antibodies reduced inflammatory hyperalgesia

    (Lewin et al., 1994; Woolf et al., 1994). NGF was later

    shown to contribute to neuropathic hyperalgesia as well

    (Herzberg et al., 1997; Theodosiou et al., 1999). The

    underlying mechanism is not clear, but it is agreed that

    NGF has a cytokine-like action on inflammatory cells

    (Otten, 1991) including mast cells, basophils, neutrophils

    and lymphocytes. Furthermore, NGF is produced and

    released by leukocytes such as macrophages and T

    lymphocytes (Vega et al., 2003). NGF is a potent degranu-

    lator of mast cells (Pearce and Thompson, 1986), which

    appear to play an important role in inflammatory (Woolf et

    al., 1996) as well as neuropathic hyperalgesia (Zuo et al.,

    2003). It seems likely that mediators released by activated

    mast cells contribute to hyperalgesia following nerve injury

    histamine has been strongly implicated (Zuo et al., 2003),

    but several other mediators may also be involved.

    1.2. Inflammatory models of neuropathic pain

    It is now apparent that early animal models of neuropathic

    pain suffer from the disadvantage that symptoms such as

    hyperalgesia are most likely the combined result of nerve

    injury itself (e.g. ectopic firing of injured sensory axons) and

    the release of algesic mediators by immune cells activated

    near the site of injury. In fact, neuropathic pain may develop

    without any injury of sensory axons. This was shown in rats

    by induction of neuropathic hyperalgesia by placing segments

    of chromic gut next to the sciatic nerve (Maves et al., 1993) or

    by lesion of the ventral roots of spinal nerves (Li et al., 2002;Sheth et al., 2002). In both cases, typical signs of neuropathic

    pain were elicited without damage to sensory axons, implying

    that sensitisation or activation of sensory fibres by inflam-

    matory mediators is sufficient to cause neuropathic pain. As a

    result of observations like these, inflammatory models of

    neuropathic pain have been developed in which carrageenan

    or complete Freund's adjuvant (Eliav et al., 1999) or zymosan

    (Chacur et al., 2001) is applied around the intact sciatic nerve

    of the rat. These models should help us to understand the role

    of inflammatory and immune mechanisms in neuropathic

    pain.

    1.3. Neuropathic pain in autoimmune diseases of thenervous system

    Autoimmune diseases of the nervous system including

    multiple sclerosis (MS) and GuillainBarr syndrome (GBS)

    are among the most common disabling neurologic diseases,

    which cause not only demyelination with impaired motor

    function, but also abnormal sensory phenomena including

    chronic neuropathic pain. MS is an autoimmune demyelinat-

    ing disease of the central nervous system that causes

    relapsing and chronic neurological impairment. Pain is

    experienced by approximately 65% of patients with MS at

    some time during the course of their disease (Kerns et al.,

    2002). GBS is an acute inflammatory demyelinating neuropa-

    thy caused by an autoimmune attack on the peripheral

    nervous system and characterised by motor disorders such

    as weakness or paralysis and variable sensory disturbances

    (Hughes et al., 1999). Pain is a common symptom of GBS

    occurring in 7090% of cases (Pentland and Donald, 1994;

    Moulin et al., 1997). Immune cells including macrophages and

    T cells are believed to play a critical role in the pathogenesis of

    both MS and GBS and may contribute to the related pain.

    Studying neuropathic pain in these autoimmune syndromes

    has been hampered by the lack of proper animal models for

    autoimmunity-related pain. However, recent study in mice

    has shown that transient demyelination of peripheral affer-

    ents without axonal loss results in neuropathic pain behav-

    iour (Wallace et al., 2003), and a suitable animal model of MS-

    related pain has recently been developed using both active

    and passive induction of experimental autoimmune enceph-

    alomyelitis (Aicher et al., 2004).

    1.4. Direct and indirect actions of mediators released by

    immune cells

    While there is good evidence that inflammatory mediators

    contribute to neuropathic pain, it is not clear just how or

    where these mediators act. Do they act directly on receptors

    located on the cell membrane of the neurone or do they

    activate non-neuronal cells (such as mast cells or Schwann

    243B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    5/25

    cells), which then release a secondary mediator that exerts a

    direct action on neuronal receptors? Are the neuronal

    receptors for chemical mediators located on the nerve

    terminals or cell bodies of sensory neurones, even though

    these may be many centimetres from the lesion site? The

    answer to the first question is that both direct and indirect

    mechanisms are likely to operate. Some mediators (such as

    prostaglandin E2) probably sensitise nociceptors directly byacting on receptors on the neuronal cell membrane (Pitchford

    and Levine, 1991). While IL-1 may also have a direct action on

    nociceptors (Sommer and Kress, 2004), it has additional

    indirect actions by eliciting the production of prostaglandins

    (Cunha et al., 1992) or even more indirectly via neural

    pathways from vagal afferents to the brainstem and then to

    the spinal cord (Watkins et al., 1995). Theanswer to thesecond

    question is that receptors for inflammatory mediators may be

    located not only on the neuronal cell body and peripheral

    nerve terminals, but also on the axon itself.

    Recent work by Grafe and his colleagues makes it clear

    that some of the neuronal receptors are located on the axon

    itself (Irnich et al., 2002; Lang et al., 2003; Moalem et al.,

    2005). This makes it easier to understand how mediators

    released by immune cells clustered around the nerve lesion

    can influence the excitability of sensory neurones since

    these mediators would be released close to axonal receptors

    and would not need to diffuse distally to nerve terminals or

    proximally to cell bodies. For example, it was shown that

    ATP increased the excitability of unmyelinated C-fibres in an

    in vitro preparation of an isolated segment of the sural

    nerve, in which no cell bodies or nerve terminals were

    present. Tissue levels of ATP are raised in inflamed tissue,

    and ATP activates nociceptors causing pain and hyperalgesia

    (Sawynok and Liu, 2003); it also plays a role in neuropathic

    pain (Barclay et al., 2002; Jarvis et al., 2002). It may well be

    that ATP contributes to neuropathic pain by its action on

    axonal receptors.

    2. Immune cells

    Several inflammatory and immune-like glial cells have been

    implicated in the pathogenesis of neuropathic pain. They

    include mast cells, neutrophils, macrophages and T cells in

    the peripheral nervous system (Fig. 1) and microglia (Fig. 2)

    and astrocytes in the central nervous system. Table 1

    summarises immune cells and the mediators they release

    with their most significant actions.

    2.1. Mast cells

    Mast cells are not only critical effector cells in allergic

    disorders but are also important initiators and effectors of

    innate immunity (Galli et al., 2005). There is a resident

    population of mast cells in the peripheral nerve (Olsson,

    1968), and these mast cells are degranulated at the site of a

    nerve lesion (Olsson, 1967; Zuo et al., 2003). The granules

    contain mediators such as histamine, proteases and cytokines

    (Metcalfe et al., 1997; Galli et al., 2005), several of which are

    capable of sensitising or activating neurones. For example,

    histamine sensitises visceral nociceptors (Koda and Mizu-

    mura, 2002). Although it elicits itch in normal skin, histamine

    application to the skin of patients with neuropathic pain did

    not evoke an itch but instead induced a severe increase in

    spontaneous burning pain (Baron et al., 2001). Activation of

    Fig. 1 Peripheral nerve injury induces activation of resident immune cells as well as recruitment of inflammatory cells to the

    nerve. Injury of a peripheral nerveinitiates an inflammatory cascade in whichmast cells residing in the nerve are the first to be

    activated. They release mediators such as histamine (hist) and TNF, which sensitise nociceptors and contribute to the

    recruitment of neutrophils and macrophages. Mediators released by neutrophils (including the chemokine MIP-1 and the

    cytokine IL-1) assist in recruitment of macrophages. Both neutrophils and macrophages in the nerve produce and secrete

    mediators such as TNF and PGE2 that can further sensitise nociceptors. Nerve injury also initiates Schwann cell

    de-differentiation and the release of several algesic mediators such as pro-inflammatory cytokines, NGF, PGE2 and ATP. These

    initial events promote the recruitment of T cells, which can secrete a variety of cytokines depending on their subtype. This

    cocktail of mediators serves as a mechanism of enhanced inflammatory response in the injured nerve and contributes to

    neuropathic pain.

    244 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    6/25

    mast cells may also cause secretion of mediators without

    overt degranulation (Theoharides and Cochrane, 2004) by

    synthesis of lipid mediators such as prostaglandin D2 or by

    transcription, translation and secretion of a wide range of

    cytokines and chemokines (Mekori and Metcalfe, 2000).

    Activation and degranulation of mast cells therefore release

    some mediators (such as serotonin in the rat) which can elicit

    hyperalgesia by a direct action on the nociceptor (Rueff and

    Dray, 1993; Sommer, 2004). Some mast cell mediators exert a

    broader range of actions by initiating an inflammatory

    cascade (Fig. 1). For example, release of histamine, leuko-

    trienes and chemokines contributes to recruitment of leuko-

    cytes including neutrophils and macrophages (Gaboury et al.,

    1995; Malaviya and Abraham, 2000). In fact, recent work in our

    laboratory suggests that degranulation of mast cells plays a

    key role in the initiation of neuropathic pain since, in rats

    subjected to partial ligation of the sciatic nerve, stabilisation of

    mast cells with cromoglycate strongly suppressed the devel-

    opment of neuropathic pain and reduced the numbers of

    neutrophils and macrophages at the lesion site (Zuo et al.,

    2003). Treatment of the nerve-injured rats with H1 and H2

    histamine receptor antagonists also alleviated neuropathic

    hyperalgesia, although to a lesser extent than stabilisation

    with cromoglycate. This suggests that nerve injury induces

    degranulation of mast cells and release of histamine, whichacts on histamine receptors on endothelial cells to induce

    recruitment of leukocytes such as neutrophils and macro-

    phages(Yamaki et al., 1998). Both of these cell types contribute

    to neuropathic pain (Liu et al., 2000b; Perkins and Tracey,

    2000). The question of how mast cells are activated by nerve

    injury is still open, but it could be brought about by increased

    levels of adenosine (Sawynok et al., 2000) or bradykinin

    (McLean et al., 2000). Other mast cell mediators are probably

    also involved in the initiation of neuropathic pain. One

    example is tryptase, a trypsin-like serine protease that is

    specific to mast cells. One of the most prominent mast cell

    mediators, it is associated with the mast cell granules and

    released by degranulation. Tryptase acts on the protease-

    activated receptor-2 (PAR-2), which is known to be present on

    primary sensory neurones and to play a role in inflammation.

    Recently, it has been shown to trigger inflammatory hyper-

    algesia and nociceptive behaviour in rats (Kawabata et al.,

    2001; Vergnolle et al., 2001).

    2.2. Neutrophils

    Neutrophils (or polymorphonuclear leukocytes) are an essen-

    tial part of the innate immune system. Their cytoplasm

    contains granules and secretory vesicles that can release a

    wide range of microbicidal effector molecules including

    bactericidal proteins and cytokines, proteinases and reactive

    oxygen species (Witko-Sarsat et al., 2000; Faurschou and

    Borregaard, 2003). The first steps in migration towards an

    inflammatory focus areadhesionto thesurface of thevascular

    endothelium (mediated by selectins) followed by rolling and

    transendothelial migration. Neutrophils then migrate towards

    the inflammatory site up a gradient of chemoattractants.

    These include formyl peptides released by bacteria or dying

    cells and chemokines released by immune cells (Witko-Sarsat

    et al., 2000). Neutrophils contribute to inflammatory hyper-

    algesia (Levine et al., 1984, 1985; Bennett et al., 1998b) by

    release of the 15-lipoxygenase product 8R, 15S-diHETE (Levine

    et al., 1985; White et al., 1990); their release of cytokines such

    as TNF (Witko-Sarsat et al., 2000) may also contribute.

    Neutrophils are not observed in normal nerves but are found

    in significant numbers at the site of injury in injured

    peripheral nerves (Perry et al., 1987; Clatworthy et al., 1995;

    Perkins and Tracey, 2000; Zuo et al., 2003). Work in our

    laboratory showed that depletion of circulating neutrophils at

    the time of nerve injury significantly attenuated the induction

    of hyperalgesia. However, depletion of neutrophils at day

    8 post-injury did not alleviate hyperalgesia after its normal

    induction since by this time neutrophil numbers at the site of

    nerve injury have declined to negligible levels(Zuo etal.,2003).

    It seems likely that nerve injury induces an inflammatory

    cascade initiated by mast cell activation. Activated mast cells

    release mediators that promote migration of neutrophils from

    Fig. 2 Peripheral nerve injury induces glial activation in the

    dorsal horn of the spinal cord. Injury to a peripheral nerve

    initiates increased release of neurotransmitters such as

    glutamate, substance P and possibly ATP from the central

    terminals of primary afferents. These neurotransmitters can

    activate both second order neurones and glia. For example,

    ATP can activate glial cells through binding to their P2X4receptors, and fractalkine released by second order neurones

    can activate glia through binding to their CX3CR1 receptors.

    The TLR4 receptor is involved in glial activation, but the

    nature of its specific ligand in the spinal cord is unclear.These triggers cause production and release of inflammatory

    mediators including pro-inflammatory cytokines (such as

    TNF and IL-1), glutamate (Glu), prostaglandins (PGs) and

    nitric oxide (NO) from glial cells. These agents are then

    capable of further enhancing glial activation and production

    of inflammatory mediators that sensitise dorsal horn

    neurones, thereby contributing to neuropathic pain.

    245B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    7/25

    Table 1 Immune cells, mediators released and actions

    Cell type Mediators Actions

    Mast cells Cytokines and

    chemokines (Galli

    et al., 2005; Mekori

    and Metcalfe, 2000)

    Recruit leukocytes;

    IL-1 facilitates the

    release of CGRP from

    nociceptors (Fukuoka

    et al., 1994) and elicits

    hyperalgesia (Ferreira

    et al., 1988; Follenfant

    et al., 1989; Sung et al.,

    2004; Zelenka et al.,

    2005); TNF sensitises

    C-fibres ( Junger and

    Sorkin, 2000; Sorkin

    et al., 1997) and DRG

    neurones (Schfers et

    al., 2003a,b) and

    produces hyperalgesia

    (Cunha et al., 1992;

    Schfers et al., 2003a,b;

    Zelenka et al., 2005).

    Chemokines produce

    excitatory effects on

    DRG neurons and

    produce allodynia

    (Oh et al., 2001)

    Histamine

    (Mekori and

    Metcalfe, 2000)

    Sensitises nociceptors

    (Koda and Mizumura,

    2002), recruits

    leukocytes (Gaboury

    et al., 1995; Yamaki

    et al., 1998); enhances

    neuropathic pain in

    patients (Baron et al.,

    2001)

    Leukotriene B4

    (Galli et al., 2005;

    Mekori and

    Metcalfe, 2000)

    Recruits neutrophils

    (Bennett et al.,

    1998a,b; Levine et al.,

    1984)

    Mast cell protease

    (e.g. tryptase)

    (Metcalfe et al.,

    1997)

    Activates PAR-2

    receptor (Kawabata

    et al., 2001; Vergnolle

    et al., 2001)

    Nerve growth

    factor (Bonini et

    al., 2003)

    Sensitises nociceptors

    (Bennett et al.,

    1998a,b; Bennett,

    2001; Koltzenburg et

    al., 1999a,b), indirectactions via immune

    cells and sympathetic

    neurones (Bennett et

    al., 1998a,b; Lewin et

    al., 1994; Woolf et al.,

    1996)

    Prostaglandins

    (PGD2, PGE2) (Galli

    et al., 2005; Mekori

    and Metcalfe, 2000)

    PGE2 sensitises

    nociceptors (Taiwo

    and Levine, 1989),

    depolarises spinal

    neurons (Baba et al.,

    2001)

    Table 1 (continued)

    Cell type Mediators Actions

    Serotonin (in

    murine rodents)

    (Galli et al., 2005)

    Sensitises nociceptors

    (Lang et al., 1990;

    Rueff and Dray, 1993)

    and sensory axons

    (Moalem et al., 2005)

    Neutrophils 8R, 15S-diHETE

    (Levine et al., 1986)

    Sensitises nociceptors

    (White et al., 1990)

    Cytokines

    (Witko-Sarsat et al.,

    2000) and chemokines

    (Scapini et al., 2000)

    See above entry for

    cytokines and

    chemokines

    Defensins (Faurschou

    and Borregaard, 2003)

    Recruit monocytes,

    T-cells (Faurschou and

    Borregaard, 2003;

    Welling et al., 1998)

    Reactive oxygen

    species (Witko-Sarsat

    et al., 2000)

    Sensitise neurons in

    PNS and CNS (Aley et

    al., 1998; Kim et al.,

    2004; Liu et al., 2000a,b;

    Meller and Gebhart,

    1993; Twining et al.,

    2004)

    Opioid peptides

    (Brack et al., 2004)

    Anti-nociceptive (Brack

    et al., 2004)

    Macrophages Cytokines (Nathan,

    1987) and

    chemokines

    See above entry for

    cytokines and

    chemokines

    Prostaglandins

    (PGE2, PGI2)

    (Nathan, 1987)

    PGE2, PGI2 sensitise

    nociceptors (Taiwo and

    Levine, 1989), PGE2 acts

    on spinal neurons (Baba

    et al., 2001)

    Reactive oxygen

    species (Nathan, 1987)

    See above entry for

    reactive oxygen species

    T lymphocytes Pro-inflammatory

    cytokines (Mosmann

    et al., 1986; Sad et

    al., 1995)

    Increase

    hyperalgesia and

    allodynia (Moalem

    et al., 2004)

    Anti-inflammatory

    cytokines (Mosmann

    et al., 1986; Sad et al.,

    1995)

    Decrease hyperalgesia

    and allodynia (Moalem

    et al., 2004)

    Glia and

    Schwann

    cells

    Cytokines and

    chemokines

    (Hanisch, 2002)

    See above entry for

    cytokines and

    chemokines

    Excitatory amino

    acids (Araque et al.,

    2001; Watkins et al.,

    2001)

    Modulate synaptic

    transmission and

    neuronal excitability

    (Araque et al., 2001)

    246 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    8/25

    small blood vessels into inflamed tissue (Gaboury et al., 1995),

    and neutrophils in turn release mediators such as chemokines

    and defensins which are chemotactic for macrophages (Well-

    ing et al., 1998; Scapini et al., 2000) (Fig. 1). Finally, it is worth

    noting that neutrophils may also have an anti-inflammatory

    and anti-nociceptive role. MRP-14 is a calcium-binding protein

    that forms a significant proportion of the cytoplasmic protein

    in neutrophils. It de-activates macrophages in vitro and

    suppresses inflammatory pain in mice (Giorgi et al., 1998).

    Under inflammatory conditions, neutrophils may secrete

    opioid peptides, which bind to opioid receptors on peripheral

    sensory neurones and mediate anti-nociception (Brack et al.,

    2004). The evidence for an anti-nociceptive role for neutro-

    phils appears to contradict the pro-nociceptive role outlined

    above. However, neuropathic pain and inflammatory pain

    differ in some respects, even though they share some basic

    mechanisms. In particular, a tertiary peripheral site of opioid

    analgesia is initiated by acute inflammation which may notbe

    activated in neuropathic pain (Bridges et al., 2001). This

    difference may help to explain why opioids are less effective

    in relieving neuropathic pain than inflammatory pain (Delle-

    mijn, 1999).

    2.3. Macrophages

    Macrophages phagocytose foreign particles such as microbes

    but also have an important role in removing injured or dead

    tissue. In the context of this review, they play a vital part in

    phagocytosing the dead or dying remnants of injured

    Schwann cells and axotomised axons in Wallerian degener-

    ation (Brck, 1997). There is a resident population of

    macrophages in the peripheral nerve and dorsal root ganglia;

    their equivalent in the CNS is the microglia. In peripheral

    nerve, the resident macrophages are assisted with clearance

    of cellular debris by an influx of haematogenous macro-

    phages (Bendszus and Stoll, 2003; Mueller et al., 2003).

    Clearance of debris is a prerequisite to regeneration in

    peripheral nerves (Perry and Brown, 1992; Correale and Villa,

    2004), and Wallerian degeneration is delayed in mice with a

    genetic defect which delays the recruitment of macrophages

    (Perry and Brown, 1992; Araki et al., 2004). Hyperalgesia is

    also delayed in these mice, suggesting that macrophages

    contribute to pain resulting from nerve injury (Myers et al.,

    1996). This suggestion was confirmed by the demonstration

    that depletion of macrophages in nerve-injured rats allevi-

    ated neuropathic hyperalgesia (Liu et al., 2000b). However,

    another study found only a limited role of macrophages inthe generation of mechanical allodynia following nerve

    injury (Rutkowski et al., 2000). Furthermore, in a model of

    neuropathic pain, the development of mechanical allodynia

    was totally abrogated in mice with a knockout for the

    chemokine receptor CCR2, involved in monocyte recruitment

    (Abbadie et al., 2003). It is noteworthy that macrophages also

    invade the dorsal root ganglia following peripheral nerve

    lesion and may contribute to neuropathic pain by release of

    excitatory agents that generate ectopic activity in sensory

    neurones (Hu and McLachlan, 2002).

    What are the signals that recruit macrophages? Essential

    roles are played by monocyte chemoattractant protein-1,

    MCP-1; macrophage inflammatory protein-1, MIP-1; and

    the IL-1 (Perrin et al., 2005); the latter two are known to be

    released by neutrophils (Witko-Sarsat et al., 2000). Release of

    defensin by neutrophils may also be involved (Welling et al.,

    1998). Which are the mediators that are released by macro-

    phages to cause hyperalgesia? Macrophages secrete prosta-

    glandins, including prostaglandin E2 and I2 (Nathan, 1987),

    which sensitise primary afferents directly. Prostaglandin

    release by macrophages is strongly implicated in neuropathic

    pain since inhibition of cyclooxygenase, an enzyme respon-

    sible for prostaglandin synthesis, relieves hyperalgesia in

    nerve-injured rats (Syriatowicz et al., 1999), and cyclooxygen-

    ase-2 is upregulated in macrophages in the injured nerve (Ma

    and Eisenach, 2002, 2003b). Other algesic mediators that are

    released by macrophages (Nathan, 1987) and most likely

    contribute to neuropathic pain include reactive oxygen

    species and the cytokines TNF (Sommer et al., 1998b), IL-1

    and IL-6 (Sommer and Kress, 2004) (Fig. 1).

    2.4. Lymphocytes

    Lymphocytes can be classed as B lymphocytes, responsible

    for antibody production, T lymphocytes, which are the

    mediators of cellular immunity, and natural killer cells.

    Both T cells and natural killer cells are found at the site of

    the nerve lesion in rat models of neuropathic pain (Cui et

    al., 2000). T cells are also found in increased numbers in

    the dorsal root ganglia and spinal cord after lesions of the

    peripheral nerve, leading to the suggestion that they may

    play a role in generating neuropathic pain (Hu and

    McLachlan, 2002; Sweitzer et al., 2002). This suggestion

    has now been confirmed by the finding that congenitally

    athymic nude rats, which lack mature T cells, develop

    significantly less mechanical allodynia and thermal hyper-

    algesia when subjected to sciatic nerve injury than their

    heterozygous littermates (Moalem et al., 2004). T cells are

    rather heterogeneous, they can be divided into CD4+

    (helper) and CD8+ (cytotoxic) cells, and each of these

    subpopulations can be further divided into type 1 and

    type 2 subsets according to their cytokine secretion profile

    Table 1 (continued)

    Cell type Mediators Actions

    Nitric oxide (Minghetti

    and Levi, 1998)

    Activates guanylyl

    cyclase, sensitises

    neurons in CNS (Luo

    and Cizkova, 2000;

    Minghetti and Levi,

    1998)

    Prostaglandins

    (Minghetti and Levi,

    1998; Muja and

    DeVries, 2004)

    See above entries

    for prostaglandins

    ATP (Anderson et al.,

    2004; Liu et al., 2005)

    Activates nociceptors

    (Hamilton and

    McMahon, 2000),

    increases sensory

    C-fibre excitability

    (Irnich et al., 2002;

    Moalem et al., 2005)

    247B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    9/25

    (Mosmann et al., 1986; Sad et al., 1995). The functions of T

    helper 1 (Th1) and Th2 cells correlate well with their

    distinctive cytokines. Th1 cells produce IL-2 and interferon-

    gamma and are involved in cell-mediated inflammatory

    reactions, while Th2 cells produce IL-4, IL-5, IL-6, IL-9, IL-10

    and IL-13, are involved in antibody and allergic responses

    and inhibit synthesis of pro-inflammatory cytokines by Th1

    cells (Mosmann and Sad, 1996). Th1 and Th2 cytokines aremutually inhibitory for the functions of the reciprocal

    phenotype. Recent work by Moalem et al. has demonstrated

    that passive transfer of type 1 T cells, which produce pro-

    inflammatory cytokines, into nude rats increased their

    sensitivity to noxious stimuli to a level comparable with

    that of heterozygous rats. By contrast, passive transfer of

    type 2 T cells, which release anti-inflammatory cytokines,

    into heterozygous rats reduced their pain sensitivity

    (Moalem et al., 2004). These findings indicate that T cells

    take part in the course of neuropathic pain (Fig. 1) and

    suggest that Th1 and Th2 cells have opposite effects on

    neuropathic pain as a result of the distinct sets of

    cytokines they release. The role of B cells in neuropathic

    pain remains to be elucidated.

    2.5. Glia and Schwann cells

    Glial cells surround and support neurones in the nervous

    system and are 10 to 50 times as numerous as neurones. In

    the last few years, it has become clear that glia are by no

    means passive bystanders in the nervous system but have

    important metabolic and immune functions. In the periph-

    eral nervous system, glia are represented by Schwann cells,

    while, in the CNS, there are three types of glial cell that can

    be distinguished the astrocytes and oligodendrocytes

    (macroglia) and the microglia. Microglia form a population

    of resident macrophages in the CNS, constituting about 5

    10% of glial cells in the CNS. In the resting state, they have a

    small soma with fine processes, but, when activated by

    stimuli such as trauma, ischemia or inflammation, they take

    on an amoeboid shape and function as phagocytes ( Vilhardt,

    2005). At this stage, they express the major histocompatibil-

    ity complex (MHC), which has a role in presenting antigen to

    T lymphocytes, and release mediators that include pro-

    inflammatory cytokines (Piehl and Lidman, 2001). Astrocytes

    represent the largest cell population in the CNS. They closely

    interact with neurones to provide active maintenance of

    homeostasis by regulating extracellular ion and neurotrans-

    mitter concentrations and extracellular pH in their micro-

    environment. The relative contribution of microglia and

    astrocytes to neuropathic pain is still under investigation.

    However, recent studies suggest that microglia are more

    important for the initiation, while astrocytes are more

    important for the maintenance of neuropathic pain (Colburn

    et al., 1999; Raghavendra et al., 2003). In this review, we will

    use the term glia for both microglia and astrocytes in the

    CNS. Evidence has been accumulating since the early 1990s

    that spinal glia contribute to neuropathic pain (DeLeo and

    Colburn, 1999; Watkins et al., 2001; McMahon et al., 2005;

    Tsuda et al., 2005). Microglia are activated by peripheral

    nerve injury (Colburn et al., 1999; Tanga et al., 2004), and

    inhibition of microglial activation in the several models of

    neuropathic pain inhibits the development of hyperalgesia

    and allodynia (Raghavendra et al., 2003; Ledeboer et al.,

    2005). Glia are activated by several mediators including ATP,

    bradykinin, substance P and prostaglandins (Hosli and Hosli,

    1993; Watkins et al., 2001). ATP is known to activate

    nociceptors in the periphery by an action on P2X3 receptors

    (Cook et al., 1997), but, in the CNS, it activates P2X4 receptors

    that are selectively expressed by activated microglia andappear to be required for neuropathic pain (Tsuda et al.,

    2005). Recent evidence suggests that activation of microglia

    by fractalkine (Milligan et al., 2004; Verge et al., 2004) and by

    the Toll-like receptor 4 (TLR4) (Tanga et al., 2005) is also

    involved in the initiation of neuropathic pain (Fig. 2).

    Fractalkine is a chemokine expressed on the surface of

    spinal neurones, which activates the CX3CR1 receptor on

    microglia, while the Toll-like receptors recognise invariant

    molecular structures of pathogens and specific ligands

    released after nerve injury.

    Although we have some idea of how glial cells are

    activated, there is little concrete evidence on how activated

    glial cells regulate neuronal function to bring about symptoms

    of neuropathic pain. Activated glia release a number of

    mediators that excite spinal neurones (Watkins et al., 2001),

    including prostanoids and nitric oxide (Minghetti and Levi,

    1998), cytokines and chemokines (Hanisch, 2002), excitatory

    amino acids (Araque et al., 2001) and ATP (Anderson et al.,

    2004). However, the role and relative importance of these glial

    mediators in neuropathic pain still need to be established.

    Interestingly, microglia have been recently implicated in

    mirror-image pain, pain arising from sites contralateral to

    the site of pathology (Koltzenburg et al., 1999b; Twining et al.,

    2004). The mechanisms underlying the expansion of pain to

    the contralateral site are not clear. However, one possible

    mechanism is through spinal gap junction activation. Recent

    study has demonstrated that treatment with a gap junction

    decoupler reverses neuropathy-induced mirror-image pain

    (Spataro et al., 2004). Thus, strong activation of glia at one site

    can lead to gap junctional propagation of calcium waves that

    activates distant glia, leading to release of pain-enhancing

    substances.

    Schwann cells in the peripheral nervous system provide

    the myelin sheath in myelinated axons and also envelop

    small bundles of unmyelinated axons (Remak bundles). The

    primary role of Schwann cells was long thought to be the

    provision of an insulating sheath around myelinated axons

    (facilitating conduction), maintenance of suitable environ-

    ment for peripheral axons, and guiding regenerating axons

    back to their targets after injury. In recent years, an immune

    function for Schwann cells has received increasing recogni-

    tion since they interact with the immune system in T-cell-

    mediated immune responses by expressing MHC class II

    molecules (Bergsteinsdottir et al., 1992). Schwann cells

    release several algesic mediators, including TNF (Murwani

    et al., 1996; Wagner and Myers, 1996a; Shamash et al., 2002 ),

    IL-1 (Bergsteinsdottir et al., 1991; Shamash et al., 2002), IL-6

    (Bolin et al., 1995), NGF (Matsuoka et al., 1991), prostaglandin

    E2 (Muja and DeVries, 2004) and ATP (Liu et al., 2005).

    Schwann cells also express a number of ion channels and

    receptors for mediators including glutamate (Liu and Ben-

    nett, 2003), ATP (Grafe et al., 1999; Irnich et al., 2001; Liu et

    248 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    10/25

    al., 2005) and IL-1 (Skundric et al., 1997). Given the close

    relationship between Schwann cells and peripheral axons

    and the presence of axonal receptors for mediators such as

    ATP (Irnich et al., 2001, 2002), it seems very likely that

    Schwann cells contribute to neuropathic pain by sensitising

    or activating the axons of nociceptors (Fig. 1). However, it

    has not yet been possible to obtain direct evidence for this

    as selective inhibition or depletion of Schwann cells in anintact animal has not yet been achieved.

    3. Mediators

    The mediators released by inflammatory and immune cells

    may act directly to sensitise or activate neurones (usually

    nociceptors in the periphery or dorsal horn neurones in the

    spinal cord). Alternatively, they may act on a non-neuronal

    cell, which on activation releases another mediator that does

    act directly on the neurone. These mediators form a long and

    increasing list that includes bradykinin, ATP and adenosine,serotonin, eicosanoids, cytokines, neurotrophins and reactive

    oxygen species (Dray, 1995). This list is not exhaustive, and

    some inflammatory mediators such as protons and histamine

    (see the Mast cells section) are not reviewed here.

    3.1. Bradykinin

    Bradykinin and kallidin are peptides that are formed from

    precursors in the blood and other tissues. In the blood,

    bradykinin is formed from high molecular weight kininogen

    as part of the clotting cascade, while in other tissues kallidin

    (lysyl-bradykinin) is formed from a low molecular weight

    kininogen. At least two subtypes of bradykinin receptors (B1

    and B2) have been characterised on the basis of in vivo and in

    vitro functional studies, using selective agonists and antago-

    nists of these receptors (Regoli et al., 1998). Bradykinin

    produces pain hypersensitivity by sensitising the peripheral

    terminals of nociceptors and by potentiating glutamatergic

    synaptic transmission in the spinal cord (Wang et al., 2005).

    Bradykinin also sensitises nociceptors by disinhibition of the

    TRPV1 receptor (Stucky et al., 1998; Di Marzo et al., 2002). B2

    receptors are constitutive and are found on a number of cell

    types including sensory neurones and microglia (Dray, 1997).

    Increased expression of B1 receptors plays a prominent role in

    inflammatory hyperalgesia (Dray and Perkins, 1993; Dray,

    1997), and this upregulation is facilitated by cytokines such as

    IL-1 (Marceau, 1995). Bradykinin acts mainly on the B2

    receptor, while des-Arg9-bradykinin, the major metabolite of

    bradykinin, has high affinity for the B1 receptor but little

    affinity for the B2 receptor (Dray and Perkins, 1993; Khasar et

    al., 1995). Both B1 and B2 receptors have been shown to be

    involved in peripheral inflammatory hyperalgesia (Dray and

    Perkins, 1993; Dray, 1997). Bradykinin stimulates macrophages

    to release TNF and IL-1 (Tiffany and Burch, 1989), as well as

    factors that are chemotactic for neutrophils and monocytes

    (Sato et al., 1996). Bradykinin also elicits the release of

    histamine from mast cells but does so by non-specific binding

    tothe cell surface rather than toB1 or B2receptors (Reissmann

    et al., 2000). In fact, peptide antagonists of B1 or B2 receptors

    may also bind non-specifically to the cell surface and elicit

    degranulation (Griesbacher and Rainer, 1999).

    Injury of the sciatic nerve in rats led to upregulation of B2

    and B1 receptors on lumbar dorsal root ganglia (Eckert et al.,

    1999; Levy and Zochodne, 2000), and in rats with a chronic

    constriction injury of the sciatic nerve, constant infusion of B2

    or B1 receptor antagonists reduced hyperalgesia (Yamaguchi-

    Sase et al., 2003). A recent study in nerve-injured mice foundthat B2 receptors were mainly expressed by the cell bodies of

    unmyelinated axons and that their expression was drastically

    decreased after nerve injury. By contrast, B1 receptors were

    newly expressed on the cell bodies of myelinated axons,

    suggesting that nerve injury induced a switch in the type of

    sensory neurone and the type of bradykinin receptor involved

    in nociception (Rashid et al., 2004). Bradykinin and its B2

    receptor have recently been implicated in central pain

    transmission. This work showed that the B2 receptor is

    expressed by dorsal horn neurones and is involved in central

    sensitisation evoked by primary afferents (Wang et al., 2005).

    Activation of B2 receptors on glial cells in the CNS could play a

    role in neuropathic pain, butthere isno evidencefor this asyet.

    3.2. ATP and adenosine

    Purines such as ATP and adenosine are well known for their

    role in energy metabolism but also have widespread effects on

    neurones and immune cells. ATP is a classical neurotrans-

    mitter but is also released by non-neuronal cells and from

    disrupted cells in injured tissue (Cook and McCleskey, 2002).

    Adenosine is a neuromodulator that is produced in the

    extracellular space by degradation of extracellular ATP and

    is also transported from the cytoplasm into the interstitial

    space by transport proteins (Linden, 1994). Inosine is produced

    by deamination of adenosine. It mayreach millimolar levelsin

    ischemic tissue and can degranulate mast cells ( Jin et al.,

    1997).

    In 1977, Bleehen and Keele reported that ATP, ADP, AMP

    andadenosine allproducedpain when applied to a blister base

    in man (Bleehen and Keele, 1977). Since then, it has been

    found that adenyl compounds and other purines activate

    nociceptors and elicit pain by their actions on purinergic

    receptors (Burnstock and Wood, 1996; Sawynok, 1998; Hamil-

    ton and McMahon, 2000). Receptors for purines have been

    classified into adenosine (P1) receptors and nucleotide (P2)

    receptors (Ralevic and Burnstock, 1998). P2 receptors can be

    subdivided into P2X receptors, which are ATP-gated ion

    channels (Chizh and Illes, 2001), and P2Y receptors, which

    are G-protein coupled receptors activated by purine or

    pyrimidine nucleotides such as ATP or UTP (von Kugelgen

    and Wetter, 2000; Burnstock and Knight, 2004).

    Initial experiments on the role of ATP in neuropathic pain

    implicated P2X3 receptors, although results were apparently

    contradictory. Immunocytochemical studies in rats and

    humans found that P2X3 receptor expression was significantly

    reduced after axotomy (Bradbury et al., 1998; Yiangou et al.,

    2000) or partial nerve ligation (Kage et al., 2002), while the

    number of P2X3 immunoreactive neurones in the DRG or

    trigeminal ganglia was increased after chronic constriction

    injury of peripheral nerves (Eriksson et al., 1998; Novakovic et

    al., 1999). This apparent discrepancy may be due to the

    249B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    11/25

    different types of nerve injury, leading to downregulation of

    P2X3 receptors in injured cells but upregulation in intact cells

    (Tsuzuki et al., 2001; Kennedy et al., 2003). Electrophysiological

    recordings from DRG neurones supported the idea that

    axotomy results in an increase in purinergic sensitivity

    (Chen et al., 2001), most likely mediated by P2X receptors

    (Zhou et al., 2001). While these results are suggestive, they do

    not directly address the issue of neuropathic pain. This wasdone by reducing the level of expression of P2X3 subunits in

    DRG cells with intrathecal administration of antisense oligo-

    nucleotides. In the partial nerve ligation model of neuropathic

    pain, this inhibited the initiation of mechanical hyperalgesia

    and reversed established hyperalgesia (Barclay et al., 2002).

    This result was confirmed using a novel non-nucleotide

    antagonist of P2X3 and P2X2/3 activation. Blocking these

    receptors in the chronic constriction model of neuropathic

    pain attenuated both thermal and mechanical allodynia

    (Jarvis et al., 2002).

    P2X4 and P2X7 receptors have also been implicated in

    neuropathic pain. P2X4 receptors are upregulated in spinal

    microglia following nerve injury, and pharmacological block-

    ade or inhibition of P2X4 receptors reversed the accompa-

    nying allodynia (Tsuda et al., 2003; Inoue et al., 2004). P2X7receptors are expressed by immune cells, including mast

    cells, macrophages and T lymphocytes. They have a key role

    in the secretion of IL-1. Recently, it was shown that

    disruption of the P2X7 purinoreceptor gene abolishes neuro-

    pathic pain and that the receptor is upregulated in human

    DRGs and injured nerves from neuropathic pain patients

    (Chessell et al., 2005).

    P2Y1 and P2Y2 receptors are found in sensory neurones

    including nociceptors (Molliver et al., 2002; Xiao et al., 2002;

    Stucky et al., 2004) and in Schwann cells (Mayer et al., 1998). In

    normal rats, P2Y1 mRNA is found in about 20% of DRG

    neurones(large as well as small), butthis percentage increases

    to about 70% after section of the sciatic nerve, suggesting a

    possible role in neuropathic pain (Xiao et al., 2002). Intrathecal

    administration of P2Y receptor agonists appears to have an

    inhibitory effect on neuropathic pain, acting via P2Y2 and/or

    P2Y4 receptors and P2Y6 receptors (Okada et al., 2002).

    Adenosine elicits cutaneous pain and hyperalgesia when

    applied peripherally (Bleehen and Keele, 1977; Taiwo and

    Levine, 1990a). It also activates axonal receptors on human C-

    fibres (Irnich et al., 2002; Lang et al., 2002). However, its role in

    the CNS is primarily inhibitory (see Sawynok and Liu, 2003 for

    review). Thus, in rats with spinal nerve ligation, spinal

    administration of adenosine resulted in a dose-dependent

    reduction in tactile allodynia (Lavand'homme and Eisenach,

    1999). Inhibition of adenosine kinase increases the availability

    of extracellular adenosine in the spinal cord, and spinal

    administration of adenosine kinase inhibitors relieves tactile

    allodynia or suppresses neuronal responses in nerve-injured

    rats (Suzuki et al., 2001; Zhu et al., 2001). These inhibitors may

    therefore hold some promise for the treatment of patients

    with neuropathic pain.

    3.3. Serotonin

    Serotonin (5-hydroxytryptamine, 5HT) is a neurotransmitter

    synthesised and released by neurones in the CNS. Messenger

    RNAs for 5HT1B, 5HT1D, 5HT2A,5HT2C, 5HT3 and 5HT7 receptors

    have been found in dorsal root ganglia (Pierce et al., 1996), and

    5HT receptors are also found in the cytoplasm of Schwann

    cells (Yoder et al., 1997).

    There is a large projection of serotonergic neurones from

    the raphe nuclei to laminae 1 and 2 of the spinal cord, and

    these serotonergic axons contribute to the descending inhibi-

    tion of pain (Millan, 2002). While serotonin suppressesnociception at the level of the spinal cord, it enhances it in

    the periphery, where serotonin sensitises nociceptors to

    thermal stimuli and to bradykinin application (Lang et al.,

    1990; Rueff and Dray, 1993). Serotonin also increases the

    excitability of sensory C-fibres in isolated segments of

    peripheral nerve (Moalem et al., 2005). In the periphery,

    serotonin is not released as a neurotransmitter by the axon

    terminals of serotonergic neurones but is an inflammatory

    mediator released by platelets and murine mast cells (seroto-

    nin is not released by human mast cells). When injected into

    the rat's paw, it induces a hyperalgesia thought to bedue to an

    action on 5HT1A receptors (Taiwo and Levine, 1992; Hong and

    Abbott, 1994).

    Work in our laboratory suggested that serotonin con-

    tributes to established mechanical hyperalgesia in the

    partial ligation model of neuropathic pain. Subcutaneous

    injection of a 5HT2A receptor blocker or a 5HT3 receptor

    blocker alleviated hyperalgesia in a dose-dependent fashion

    when injected into the hindpaw of the nerve-injured limb.

    Injection of the blockers into the contralateral hindpaw did

    not relieve hyperalgesia, suggesting that the effect was

    mediated locally rather than systemically (Theodosiou et

    al., 1999). Other research on the involvement of serotonin in

    neuropathic pain has focussed on its role in the descending

    inhibition of pain. Tricyclic antidepressants (TCAs) are

    widely used to treat neuropathic pain in patients and inhibit

    reuptake of monoamines such as serotonin and noradren-

    aline into nerve terminals. This is thought to increase the

    efficacy of descending monoaminergic pathways that inhibit

    pain. Some TCAs are effective in alleviating neuropathic

    pain in animal models, but selective inhibitors of serotonin

    reuptake were not effective in animal models or in patients

    (Max et al., 1992; Jett et al., 1997). However, fenfluramine,

    which not only inhibits serotonin reuptake but also elicits

    releases of serotonin from presynaptic terminals, produced a

    significant reduction in mechanical allodynia in rats with

    tight ligation of the L5/L6 spinal nerves (Wang et al., 1999).

    Serotonin reuptake is mediated by the serotonin transporter

    (5HTT), and mice deficient for the serotonin transporter did

    not develop thermal hyperalgesia following chronic constric-

    tion of the sciatic nerve (Vogel et al., 2003). Whether this was

    due to altered tissue levels of serotonin or downregulation of

    5HT receptors was not clear.

    3.4. Eicosanoids

    Arachidonic acid is a polyunsaturated fatty acid that is found

    in the cell membrane. It is the major precursor of the

    eicosanoids, and, once it has been freed from the cell

    membrane by phospholipase A2, it is converted by enzymes

    in the arachidonate cascade to compounds that include

    prostaglandins, thromboxanes and leukotrienes.

    250 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    12/25

    Arachidonic acid is metabolised to prostanoids by the

    cyclooxygenase pathway and to leukotrienes by the lipox-

    ygenase pathway (Wolfe and Horrocks, 1994; Smith et al.,

    2000). It may also be converted to isoprostanes by peroxida-

    tion; the isoprostanes are inflammatory mediators that

    augment nociception (Evans et al., 2000).

    Prostaglandins PGE2 and PGI2 induce hyperalgesia in the

    periphery (Taiwo and Levine, 1990b) where they act directlyon the terminals of nociceptors (Taiwo and Levine, 1989)

    and probably on macrophages as well (Ma and Eisenach,

    2003a). More recently, it has been established that prosta-

    glandins also contribute to nociception at the level of the

    spinal cord (Malmberg and Yaksh, 1992; Vanegas and Schaible,

    2001).

    There are two primary isoforms of cyclooxygenase, COX-1

    and COX-2. COX-1 is expressed constitutively in most tissues

    and has a homeostatic or housekeeping role, while COX-2

    expression is usually lowbut canbe induced by factors such as

    inflammatory cytokines in cells including mast cells, macro-

    phages (Smith et al., 2000) and spinal neurones (Samad et al.,

    2002). In fact, COX-1 may also be induced in some conditions

    such as spinal cord injury (Schwab et al., 2000; Samad et al.,

    2002). Prostaglandins exert their effects by actions on G-

    protein-coupled receptors including the EP receptors (EP14 for

    PGE2) and the IP receptor for PGI2 or prostacyclin (Hata and

    Breyer, 2004).

    Work in our laboratory showed that mechanical hyper-

    algesia in nerve-injured rats wasalleviated for up to 10 days by

    subcutaneous injection of indomethacin (a classic inhibitor of

    cyclooxygenase) into the affected hindpaw. Subcutaneous

    injection of selective COX-2 inhibitors or an EP1 receptor

    blocker relieved thermal as well as mechanical hyperalgesia,

    but with a shorter timecourse. We concluded that increased

    expression of prostaglandins in the region of the nerve lesion

    contributed to neuropathic pain (Syriatowicz et al., 1999). It

    was then shown in several animal modelsof neuropathic pain

    that the number of COX-2 immunoreactive cells was dramat-

    ically increased in the region of the nerve lesion, that most of

    these cells were infiltrating macrophages (Ma and Eisenach,

    2002, 2003b) and that this was the case in human patients as

    well as nerve-injured rats (Durrenberger et al., 2004). In fact,

    the increase in COX-2 expression appears to be biphasic, with

    an early increase in Schwann cells (1 day after spinal nerve

    injury) and an increase in macrophage expression at about 7

    14 days (Takahashi et al., 2004). As one might expect,

    increased levels of PGE2 are found in the injured nerves

    (Schfers et al., 2004). Furthermore, cells immunoreactive for

    EP receptors are found in the injured nerve, but not in normal

    intact nerves. Once again, many of the immunoreactive cells

    prove to be macrophages (Ma and Eisenach, 2003a). These

    observations, based on several animal models of sciatic nerve

    injury, support the idea that upregulation of COX-2 and EP

    receptorsin the injured nerve contributes to neuropathic pain.

    However, in the spared nerve injury model (Decosterd and

    Woolf, 2000), where degenerating axons distal to the lesion do

    not mingle with intact axons, treatment with the COX-2

    inhibitor rofecoxib had no effect on the development of

    allodynia and hyperalgesia (Broom et al., 2004).

    Increased expression of cyclooxygenase after nerve injury

    is not restricted to the nerve itself. COX-2 is also upregulated

    in the dorsal horn of the spinal cord and thalamus following

    peripheral nerve injury (Zhao et al., 2000), and the resulting

    allodynia is attenuated by intrathecal injection of ketorolac, a

    COX-1 preferring inhibitor (Ma et al., 2002). COX-1 is expressed

    constitutively in glial cells and motor neurones of normal rats.

    It is also upregulated after ligation of L5L6 spinal nerves so

    that the number of cells immunoreactive for COX-1 is

    increased in the superficial laminae of the dorsal horn at 4days after spinal nerve ligation and remains high for at least 2

    weeks (Zhu and Eisenach, 2003). How do increased levels of

    prostaglandins in the spinal cord mediate neuropathic pain?

    There is evidence for at least twomechanisms. Thefirst is that

    PGE2 depolarises wide dynamic range neurones in the deep

    dorsal horn (Baba et al., 2001), and the second is that PGE2 also

    blocks glycinergic inhibition of neurones in the superficial

    laminae of the dorsal horn (Ahmadi et al., 2002; Harvey et al.,

    2004).

    Given the evidence for a role of cyclooxygenases in the

    development of neuropathic pain and the efficacy of cycloox-

    ygenase inhibitors in treating inflammatory pain, it is

    surprising that COX inhibitors are relatively ineffective in the

    treatment of neuropathic pain in patients (Namaka et al.,

    2004). This may be due to the predominant role of prostaglan-

    dins in the development rather than maintenance of neuro-

    pathic pain or that higher effective concentrations of COX

    inhibitors are achieved in experimental animals than in

    patients.

    There are three mammalian lipoxygenases, which catalyse

    the insertion of oxygen at positions 5, 12 and 15 of arachidonic

    acid. These are therefore referred to as 5-, 12- and 15-

    lipoxygenases. The 5-lipoxygenases form leukotrienes, and

    leukotriene B4 (LTB4) produces hyperalgesia by eliciting the

    release of a mediator from neutrophils (Levine et al., 1984,

    1985), which was identified as the 15-lipoxygenase product

    (8R, 15S)-dihydroxyeicosa-(5E-9,11,13Z)-tetraenoic acid (8R,

    15S-diHETE) (Levine et al., 1986). This was shown to sensitise

    nociceptors in the rat and may contribute to the role of

    neutrophils in neuropathic pain (see the Neutrophils section).

    Nerve growth factor elicits hyperalgesia when injected into

    the rat paw, and inhibition of 5-lipoxygenase inhibits release

    of LTB4, accumulation of neutrophils and the associated

    hyperalgesia (Amann et al., 1996; Bennett et al., 1998b). It is

    likely that NGF produces hyperalgesia by inducing the release

    of LTB4 from mast cells, leading in turn to recruitment of

    neutrophils (Bennett et al., 1998b), which then release algesic

    mediators and chemokines. Neutrophils and nerve growth

    factor have both been directly implicated in the genesis of

    neuropathic pain (Herzberget al., 1997; Theodosiou et al., 1999;

    Perkins and Tracey, 2000), and so it is likely that LTB4 and 8R,

    15S-diHETE are also involved.

    3.5. Cytokines

    Cytokines are small proteins that mediate interactions

    between cells over relatively short distances. They are mostly

    involved in responses to disease or infection. Many of them

    are referred to as interleukins, denoting a mediator released

    by one leukocyte and acting on another, but they are

    synthesised by most cell types. Several are pro-inflammato-

    ry, such as IL-1, IL-6 and TNF, while others such as IL-10 are

    251B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    13/25

    anti-inflammatory. Exogenous administration of these pro-

    inflammatory cytokines elicits pain and hyperalgesia (Som-

    mer and Kress, 2004), but they do not appear to be involved

    in normal pain (Watkins et al., 2001). In fact, these three

    pro-inflammatory cytokines induce the production of each

    other, and they act synergistically (Watkins et al., 1999). This

    leads to the possibility of positive feedback, which could lead

    to chronic pain if not appropriately regulated. Other cyto-kines such as IL-4 and IL-10 are anti-inflammatory and

    suppress genes that code for IL-1, TNF and chemokines. The

    algesic effects of pro-inflammatory cytokines are often

    indirect, so that they may not act directly on the nociceptor

    but instead induce the expression of agents (such as PGE2)

    that themselves sensitise nociceptors. In inflammation and

    Wallerian degeneration, cytokine production is organised

    into a sequence with TNF playing a leading role (Shamash et

    al., 2002; Cunha et al., 2005). This sequence appears to be one

    of the mechanisms underlying neuropathic pain (for reviews,

    see DeLeo and Colburn, 1999; Sommer, 2001; Sorkin, 2002;

    Sommer and Kress, 2004). Chemokines are small chemotactic

    cytokines that are important for leukocyte migration and

    recruitment to damaged sites. Recently, they have been

    shown to contribute directly to nociception by producing

    excitatory effects on DRG neurones and inducing allodynia

    after injection into the rat's paw (Oh et al., 2001). The

    contribution of chemokines to pain has been reviewed

    recently (Abbadie, 2005; White et al., 2005).

    3.5.1. Interleukin-1

    Binding of IL-1 to its receptor IL1-RI on the cell surface

    initiates several signalling events, such as translocation of NF-

    B into the nucleus (Dinarello, 1999). NF-B then upregulates

    transcription of several genes, including COX-2, inducible

    nitric oxide synthase, TNF, IL-1 and IL-6 (Pahl, 1999; Tegeder

    et al., 2004). IL-1 may act directly as well as indirectly on

    nociceptors. Thus, Fukuoka and colleagues found that IL-1

    facilitated release of CGRP from nociceptors in an in vitro

    nerve-skin preparation. The latency of the effectwas too short

    for upregulation of receptors or changes in gene expression,

    and the cell bodies were not present, suggesting a direct

    sensitisation of nociceptors (Fukuoka et al., 1994; Sommer and

    Kress, 2004). IL-1 elicits hyperalgesia when injected periph-

    erally into the rat paw (Ferreira et al., 1988; Follenfant et al.,

    1989), intraneurally into rat sciatic nerve (Zelenka et al., 2005),

    intrathecally in the rat spinal cord (Sung et al., 2004) or

    centrally into various regions of the brain (see Bianchi et al.,

    1998 for review). In fact, Watkins and her colleagues have

    shown that IL-1 may induce an illness hyperalgesia via a

    kind of supraspinal loop involving sensory fibres in the

    vagus nerve, the nucleus of the solitary tract, nucleus raphe

    magnus and projections to the spinal cord (Watkins et al.,

    1994; Watkins and Maier, 1999). IL-1 is implicated in

    neuropathic pain since IL-1 and IL-1 are both upregulated

    in injured peripheral nerve (Gillen et al., 1998; Okamoto et

    al., 2001; Shamash et al., 2002). Neuropathic pain is

    alleviated in nerve-injured mice by administration of neu-

    tralizing antibodies to the IL-1 receptor (Sommer et al., 1999;

    Schfers et al., 2001) and in nerve-injured rats by intrathecal

    IL-1 receptor antagonist in combination with soluble TNF

    receptor (Sweitzer et al., 2001).

    3.5.2. Interleukin-6

    IL-6 is synthesised by many cell types, including mast cells,

    monocytes, lymphocytes, neurones and glial cells. Once IL-6

    has bound to its receptor IL-6R, it initiates two major

    intracellular cascadesone involving Janus kinases and

    STAT factors, the other using the Ras-dependent MAP kinase

    pathway (De Jongh et al., 2003). Injury of the sciatic nerve

    induces upregulation of IL-6 and its receptor in the region ofthe lesion, where it appears in macrophages and Schwann

    cells (Bolin et al., 1995; Kurek et al., 1996; Ito et al., 1998; Grothe

    et al., 2000). Upregulation of IL-6 in these macrophages

    appears to be induced by PGE2 (Ma and Quirion, 2005). Sciatic

    nerve injury also increases IL-6 levels in the DRG and spinal

    cord, particularly in the superficial laminae of the dorsal horn

    (Murphy et al., 1995; DeLeo et al., 1996; Lee et al., 2004).

    Exogenous IL-6 induces thermal hyperalgesia when injected

    into the lateral cerebral ventricles of the rat (Oka et al., 1995)

    and increases the heat-evoked release of CGRP from cutane-

    ous nociceptors (Opree and Kress, 2000; Obreja et al., 2002).

    However, there is little evidence that IL-6 plays a role in

    normal nociception. One study did report that intraplantar

    injection of IL-6 induced mechanical hyperalgesia in naiverats

    (Cunha et al., 1992). However, this finding has not been

    reproduced and later work found that intraplantar IL-6 had no

    effect on mechanical thresholds (Czlonkowski et al., 1993) or

    produced a thermal hypoalgesia (Flatters et al., 2004). In fact,

    peripheral IL-6 inhibitedheat responses of nociceptors in an in

    vitro preparation.

    There is limited evidence that IL-6 contributes to the

    development of neuropathic pain. In the in vivo spinal cord,

    peripheral IL-6 inhibited the responses of dorsal horn neu-

    rones to thermal and mechanical stimuli; after spinal nerve

    ligation, peripheral IL-6 released the inhibition of mechanical

    responses (Flatters et al., 2004). In IL-6 knockout mice,

    nociceptive responses to thermal and mechanical stimuli are

    the same as in wild-type mice (Bianchi et al., 1999; Murphy et

    al., 1999), but the development of mechanical allodynia after

    spinalnervelesion is delayed in IL-6 knockoutsby comparison

    with wild-type mice (Ramer et al., 1998). It is not clear whether

    thermal hyperalgesia induced by a spinal nerve lesion is

    reduced in IL-6 knockouts (Murphy et al., 1999) or not (Ramer

    et al., 1998).

    3.5.3. Tumour necrosis factor

    Tumour necrosis factor (TNF, TNFSF2, formerly TNF) is a

    member of a large superfamily of proteins, which have an

    unusual trifold symmetry. There is an equally large super-

    family of receptors; the receptors activated by TNF are the

    constitutively expressed TNFR1 (TNFRSF1A, p55-R) and the

    inducible TNFR2 (p75-R) (Locksley et al., 2001). TNFR1 is linked

    to pathways for cell death, whereas TNFR2 is not. However,

    activation of either receptor results in p38 MAP kinase

    signalling (Schfers et al., 2003b), translocation of NFB to

    the nucleus and activation of COX-2-dependent prostanoid

    release, as described above for IL-1 (Dinarello, 1999). TNF is

    constitutively expressed in cutaneous mast cells (Walsh et al.,

    1991), but, in injury or inflammation, it may be released by

    other cells including neutrophils and macrophages. Intraplan-

    tar injection of TNF into the rat's paw induces mechanical

    hyperalgesia (Cunha et al., 1992). This can be attributed to

    252 B R A I N R E S E A R C H R E V I E W S 5 1 ( 2 0 0 6 ) 2 4 0 2 6 4

  • 8/14/2019 Immune and inflammatory mechanism in neuropathic pain

    14/25

    sensitisation of C-fibres since subcutaneous injection in the

    distribution of the sural nerve lowered the thresholds of

    cutaneous C-fibres to mechanical stimulation and led to

    ongoing activity in some of these fibres ( Junger and Sorkin,

    2000). In fact, topical application of TNF to a restricted portion

    of the sciatic nerve led to ectopic firing of A- and C-fibres

    (Sorkin et al., 1997), and intraneural injection of TNF into rat

    sciatic nerve at physiological doses induced thermal hyper-algesia and mechanical allodynia (Zelenka et al., 2005),

    suggesting the presence of TNF receptors on the axons. An

    alternative possibility is insertion of the TNF trimer into the

    cell membrane of the axon (Baldwin et al., 1996), creating a

    pore that is permeable to sodium ions and may be voltage-

    dependent (Kagan et al., 1992).

    Injury of the sciatic nerve leads to upregulation of TNF and

    its receptors in the nerve (George et al., 1999; Shubayev and

    Myers, 2000; George et al., 2005); this upregulation is found

    mainly in Schwann cells and endothelial cells (Wagner and

    Myers, 1996a). Nerve injury also leads to increased TNF

    expression in the dorsal horn of the spinal cord and in the

    locus coeruleus and hippocampus (Ignatowski et al., 1999). An

    important role for TNF in neuropathic pain is indicated by

    reducing TNF levels in nerve-injured rats or mice (Sommer,

    2001). Inhibiting TNF synthesis with thalidomide or treatment

    with anti-TNF neutralising antibodies at the time of nerve

    injury blocked the development of hyperalgesia and allodynia

    in these animal models (Sommer et al., 1998a,b, 2001a). Treat-

    ment with etanercept, a recombinant TNF receptor (p75)-Fc

    fusion protein that acts as a TNF antagonist, reversed estab-

    lished hyperalgesia in mice with a chronic constriction injury

    of the sciatic nerve (Sommer et al., 2001b), and endoneurial

    injection of neutralising antibodies against TNF receptors

    showed that neuropathic hyperalgesia is dependent on the

    TNFR1 receptor (Sommer et al., 1998b). Furthermore, applica-

    tion of exogenous TNF to the DRG increased the sensitivity of

    injured and adjacent uninjured primary sensory neurones and

    elicited faster onset of allodynia and spontaneous pain

    behaviour after spinal nerve ligation (Schfers et al., 2003a).

    3.5.4. Interleukin-10

    IL-10 is generally regarded as anti-inflammatory. Its expres-

    sion increases gradually over at least 6 weeks following nerve

    injury (Okamoto et al., 2001), and treatment with a single dose

    of IL-10 at the site of a chronic constriction injury significantly

    reduces hyperalgesia, probably due in part to suppression of

    TNF expression and macrophage recruitment (Wagner et al.,

    1998). These results are consistent with findings that thalid-

    omide (an inhibitor of TNF synthesis) decreases TNF levels,

    increases endoneurial IL-10 levels, and alleviates hyperalgesia

    in rats with a CCI nerve lesion (Sommer et al., 1998a; George et

    al., 2000). Recent work confirms theefficacyof IL-10 in alleviat-

    ing neuropathic pain, using virally driven spinal production of

    IL-10 in neuropathic pain models (Milligan et al., 2005a,b).

    3.6. Neurotrophins

    The neurotrophins are dimeric proteins that are essential for

    the normal development of the vertebrate nervous system.

    This family includes nerve growth factor (NGF), brain-derived

    neurotrophic factor (BDNF), neurotrophin (NT)-3 and NT-4/5.

    Glial-cell-line-derived neurotrophic factor (GDNF) also has

    neurotrophic properties but is structurally unrelated to the

    dimeric neurotrophin family (Lewin and Barde, 1996).

    The neurotrophins act on a trio of tyrosine kinase (Trk)

    receptors U TrkA, TrkB and TrkC, which primarily bind NGF,

    BDNF and NT-4/5, and NT-3 respectively. There is also a low

    affinity or pan-neurotrophin receptor, p75NTR, which acti-

    vates a separate set of signalling pathways that interact withthose activated by Trk receptors (Huang and Reichardt, 2003;

    Nykjaer et al., 2005). During embryonic development, all

    small-diameter sensory neurones express TrkA, the high

    affinity receptor for NGF. Most of these neurones will become

    nociceptors, with thinly myelinated axons (A fibres) or

    unmyelinated axons (C-fibres). After birth, the expression of

    TrkA is downregulated so that TrkA is only expressed by those

    small sensory neurones which are peptidergic, whereas the

    non-peptidergic remainder (IB4 binding neurones) now

    expresses receptor components for another neurotrophin,

    GDNF (Bennett, 2001).

    Neurotrophins are synthesised and released by a several

    cell types of immune cells, including mast cells and lympho-

    cytes (Moalem et al., 2000; Bonini et al., 2003). BDNF is

    constitutively expressed by peptidergic nociceptors (Thomp-

    son et al., 1999).

    Nerve injury produces marked changes in expression of

    neurotrophins and their receptors, primarily in Schwann cells

    (Frostick et al., 1998). Schwann cells in the intact peripheral

    nerve synthesise NGF, BDNF and GDNF, and this synthesis is

    dramatically upregulated by nerve injury (Lindholm et al.,

    1987; Meyer et al., 1992; Hammarberg et al., 1996). BDNF

    synthesis is also upregulated in sensory neurones following

    axotomy (Tonra et al., 1998). In the intactsciatic nerve, mRNAs

    for the neurotrophin receptors TrkB and TrkC are expressed,

    but TrkA and p75NTR mRNAs are undetectable. Levels of

    p75NTR mRNA are markedly increased in Schwann cells after

    nerve injury, but TrkA levels remain undetectable (Funakoshi

    et al., 1993).

    It is now recognised thatsome neurotrophins (in particular,

    NGF and BDNF) play a significant role in nociception (Bennett,

    2001) so that NGF sensitises nociceptors in the periphery

    (Koltzenburg et al., 1999a) while BDNF increases the respon-

    siveness of dorsal horn neurones in the spinal cord (Pezet et

    al., 2002). Intraplantar or systemic injection of NGF induces

    hyperalgesia (Lewin et al., 1994; Andreev et al., 1995; Theodo-

    siou et al., 1999). Tissue levels of NGF are increased by

    inflammation, and the hyperalgesia resulting from inflamma-

    tion is prevented by administration of anti-NGF neutralising

    antibodies or a TrkA-IgG fusion molecule (Woolf et al., 1994;

    McMahon et al., 1995). Some of the hyperalgesic action of NGF

    is exerted directly on sensory neurones, but indirect actions

    via mast cells, neutrophils and sympa