Mol Neurobiol DOI 10.1007/s12035-014-8706-9

Peripheral Nerve Injury Modulates Neurotrophin Signaling in the Peripheral and Central Nervous System Mette Richner & Maj Ulrichsen & Siri Lander Elmegaard & Ruthe Dieu & Lone Tjener Pallesen & Christian Bjerggaard Vaegter

Received: 23 December 2013 / Accepted: 1 April 2014 # Springer Science+Business Media New York 2014

Abstract Peripheral nerve injury disrupts the normal functions of sensory and motor neurons by damaging the integrity of axons and Schwann cells. In contrast to the central nervous system, the peripheral nervous system possesses a considerable capacity for regrowth, but regeneration is far from complete and functional recovery rarely returns to pre-injury levels. During development, the peripheral nervous system strongly depends upon trophic stimulation for neuronal differentiation, growth and maturation. The perhaps most important group of trophic substances in this context is the neurotrophins (NGF, BDNF, NT-3 and NT-4/5), which signal in a complex spatial and timely manner via the two structurally unrelated p75NTR and tropomyosin receptor kinase (TrkA, Trk-B and Trk-C) receptors. Damage to the adult peripheral nerves induces cellular mechanisms resembling those active during development, resulting in a rapid and robust increase in the synthesis of neurotrophins in neurons and Schwann cells, guiding and supporting regeneration. Furthermore, the injury induces neurotrophin-mediated changes in the dorsal root ganglia and in the spinal cord, which affect the modulation of afferent sensory signaling and eventually may contribute to the development of neuropathic pain. The focus of this review is on the expression patterns of neurotrophins and their receptors in neurons and glial cells of the peripheral nervous system and the spinal cord. Furthermore, injury-induced changes of expression patterns and the functional consequences in relation to axonal growth and remyelination as well as to neuropathic pain development will be reviewed. M. Richner : M. Ulrichsen : S. L. Elmegaard : R. Dieu : L. T. Pallesen : C. B. Vaegter (*) Danish Research Institute of Translational Neuroscience DANDRITE, Nordic EMBL Partnership, and Lundbeck Foundation Research Center MIND, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000 Aarhus C, Denmark e-mail: [email protected]

Keywords Neurotrophins . Peripheral nerve injury . Regeneration . Neuropathic pain

Introduction — Nerve Injury, Neurotrophins and Their Receptors Peripheral neuropathy is a condition where damage resulting from mechanical or pathological mechanisms is inflicted on nerves within the peripheral nervous system (PNS). Physical injury is the most common cause and may result in nerves being partially or completely severed, crushed, compressed or stretched. In contrast to the central nervous system (CNS), the PNS possesses a unique ability to regenerate, recognized since ancient times and initially described by surgeons and later methodologically investigated by modern neurobiologists including Camillo Golgi and Santiago Ramon y Cajal [1]. However, despite the considerable capacity for regrowth upon injury, PNS regeneration is far from complete and functional recovery rarely returns to pre-injury levels. Several biological factors influence the communication between peripheral nerves and tissues following injury including a variety of cytokines and growth factors. Similar conditions can be observed during the development of the nervous system where the PNS strongly depends on trophic stimulation for survival, differentiation, sprouting and maturation. As damage to the adult PNS induces cellular mechanisms that control neuronal differentiation as well as growth and connectivity during development, studies of neurotrophic factors are considered to represent one of the most promising areas of research aimed at finding new, more effective treatments for peripheral neuropathies [2]. Various families of neurotrophic factors are being studied; however, signaling by the neurotrophins is probably receiving most attention. The family of neurotrophins consists of nerve growth factor (NGF), brain derived neurotrophic factor

Mol Neurobiol

(BDNF), neurotrophin-3 (NT-3) and neurotrophin-4/5 (NT-4/ 5), which can bind to two structurally unrelated receptors: the neurotrophin receptor p75NTR and the tropomyosin receptor kinases (TrkA, -B, and -C). Binding of neurotrophins to the Trk receptors is selective with NGF binding to TrkA, BDNF and NT-4/5 to TrkB and NT-3 to TrkC [3]. However, many neurons co-express Trk receptors and p75NTR, which together strengthen the affinity and specificity of the neurotrophins for the Trk receptors probably through the formation of a receptor complex [4–8]. Neurotrophins are synthesized as precursor proteins (proneurotrophins), which can be cleaved by furin or other proconvertases to produce the mature proteins of about 13– 15 kDa in size. Both pro-neurotrophins and neurotrophins are released as stable, non-covalent homodimers and are believed to elicit different signaling properties. The neurotrophins bind to their respective Trk receptors to mediate survival and differentiation via extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K) and phospholipase C-γ (PLC-γ) pathways. Neurotrophins can also provide survival signaling via p75NTR and the NF-κB pathway or affect cytoskeletal organization and neurite outgrowth via downstream RhoA kinase activity (reviewed in [3, 9]). Yet, p75NTR is better recognized for its ability to induce cell death signaling via binding of various intracellular adaptor proteins, ultimately activating the c-Jun N-terminal kinase (JNK) pathway and subsequently p53 leading to apoptosis. For many years, p75NTR-mediated apoptosis was considered to be initiated by mature neurotrophins, whereas pro-

Spinal cord dorsal horn

neurotrophins were thought to be merely inactive precursors. Increasing evidence has, however, identified important biological functions of pro-neurotrophins, one of which is the ability to induce cell death by binding to the receptor sortilin (via the pro-domain) and p75NTR (via the mature domain), thereby creating a tertiary complex that results in activation of neuronal apoptosis [10–14]. To add to the complexity, it has recently been reported that TrkA and TrkC, but not TrkB, behave as dependence receptors during development, i.e., they may also trigger neuronal death on their own in absence of activation by their corresponding neurotrophins [15]. Hence, it is apparent that the consequences of neurotrophin signaling depend on the interplay between neurotrophins/proneurotrophins and their receptors in a very complex spatial and timely manner. Injury to peripheral nerves induces dramatic changes in the expression levels of neurotrophins and their receptors in all cellular components of the PNS (neurons, satellite glial cells [SGCs] and Schwann cells) as well as in some CNS cells of the spinal cord (motor neurons and microglia). In particular, altered neurotrophin signaling is believed to be essential for the numerous complex processes underlying peripheral nerve regeneration as well as in neuropathic pain development — two important biological responses to peripheral nerve injury. In the following sections, the expression of neurotrophins and their receptors as well as injury-induced changes in expression and signaling are described in three major structural areas of the nervous system: the dorsal root ganglia (DRG), the peripheral nerves and the spinal cord dorsal horn (Fig. 1).

Dorsal rootlets

Spinal cord ventral horn

Dorsal root

Peripheral nerve injury

Ventral rootlets Dorsal root ganglion

Ventral root

Pia mater

Arachnoid mater

Peripheral nerve

Dura mater

Fig. 1 The spinal cord and the peripheral nervous system (PNS). Peripheral nerve injury induces various changes in the PNS (peripheral nerve fibers and DRGs) but also in the central nervous system (CNS) (spinal

cord). Changes include altered expression levels of neurotrophins and their receptors with concomitant altered signaling processes in both the PNS and CNS

Mol Neurobiol

each DRG fuses with the corresponding spinal ventral root at the same level of each vertebra to form a spinal nerve, which intermingles distally with other spinal nerves to form the peripheral nerves.

Dorsal Root Ganglia Anatomy at a Glance DRG neurons are primary afferent neurons responsible for transmitting sensory information from the periphery to the CNS. The DRGs are part of the PNS and are located in the neural foramen of the vertebrae, in close proximity to the CNS. The connective tissues surrounding the DRGs are continuous with the two outermost spinal meninges, the dura mater and the arachnoid mater [16]. For each cervical (C), thoracic (T), lumbar (L) and sacral (S) vertebra the DRGs are designated C, T, L or S and the corresponding number of the given vertebra in a rostro-caudal order. Thus, e.g., the DRGs of the fourth lumbar vertebra are named L4, while the second thoracic DRGs are named T2. The sciatic nerve, which is often the object in rodent nerve injury models [17], has its main part of neuronal somas in L3–L5 DRGs in mice and correspondingly in L4–L6 DRGs in rats [18]. Primary afferents have a unique pseudo-unipolar structure meaning that the soma of each neuron situated in a DRG projects one axon that bifurcates into a peripheral and a central branch. No other neuronal processes are formed by the soma (Fig. 2a). The peripheral branch extends distally towards the skin or viscera to receive sensory stimulation, whereas the central branch transmits these signals via synaptic terminals to the secondary sensory neurons of the spinal cord dorsal horns for central processing. This pseudounipolarity allows direct signal transmission from the site of stimulation in the periphery to the CNS, bypassing the neuronal soma. Even though afferent signals initiated in the periphery can bypass the soma, signal invasion to the soma provides necessary information for protein synthesis and for maintaining optimal levels of receptors and ion channels at the nerve terminals [19]. The distal root of

a

Glial Cells Glial cells are numerous in the DRGs and count Schwann cells, which wrap around the neuronal processes, and SGCs, which enclose each neuronal soma. Several SGCs, interconnected by gap junctions, together form a sheath around one neuronal soma (Fig. 2a), leaving a gap of just 20 nm between the SCGs and the neuron — the width of a synapse. This functional unit of soma and SGCs is enclosed by a layer of connective tissue [20–22]. The non-neuronal face of the slimly flattened SGCs has a basement membrane, which together with the endothelial basement membrane constitutes a blood– neuron barrier [23, 24]. Through expression of a number of receptors and channels, SGCs are believed to stabilize the neuronal microenvironment, supporting normal neuron function and providing electrical insulation (SGC protein expression reviewed in [24]). This intimate and isolated structural arrangement between SGCs and soma strongly implies that soma-SGC communication is a major determinant of neuronal activity, and that understanding injury-induced changes in this communication is important for understanding the neuronal changes and activity following peripheral nerve injury. SGCs may be compared to astrocytes of the CNS, as they have similar functions and express several similar biochemical markers, such as glutamine synthetase (GS) and, upon injury, glial fibrillary acidic protein (GFAP). However, similarity as such is confined to function, since SGCs originate from neural crest cells of the embryo, while astrocytes are derived from the neural tube [25].

b

Neuron cell body

Myelinated axons

Schwann cells

Node of Ranvier

Neuron cell body

Satellite glial cells

Satellite glial cells

Fig. 2 Dorsal root ganglia (DRG) neurons and encapsulating satellite glial cells (SGCs). a Each neuron is closely surrounded by multiple SCGs forming a neuron–SGC unit. Each pseudounipolar neuron projects one

axon that bifurcates into a branch that extends to the peripheral target tissue and a branch that extends to the spinal cord. b DRG cross-section illustrating primary sensory neurons surrounded by a single thin layer of SGCs

Mol Neurobiol

Characterization of Primary Afferent Neuron Types and Modalities A widely used system to characterize the primary afferents is according to axon size, degree of myelination and, hence, conduction velocity. The large, heavily myelinated Aα- and Aβ-fibers transmit signals of proprioception and innocuous mechanoreception, respectively. Small myelinated fibers, designated Aδ-fibers, mostly convey fast sharp pain and temperature sensation, although some Aδ-fibers convey lowthreshold mechanosensation. Small unmyelinated fibers, designated C-fibers, convey sensation of temperature, slow dull pain and itch [26–29]. DRG neurons of different modality project axons to certain laminae of the spinal cord; nociceptive afferents terminate in dorsal horn laminae I and II, whereas mechanosensitive neurons mainly terminate in laminae III and IV. Proprioceptive neurons terminate in the ventral spinal cord where they synapse with motor neurons [30]. Neuronal somas in the DRG are divided into two groups according to morphology: large, light neurons with coarsely granular cytoplasm and small, dark neurons with a denser, more uniform cytoplasm [31–34]. The large, light neurons are referred to as type A neurons and the small, dark neurons as type B neurons [35, 36]. In rats, approximately 40 % of DRG neurons are type A neurons and approximately 60 % are type B neurons, while a small percentage of medium-sized neurons fall out of these categories [37–39]. The type A and B neurons can further be subdivided according to biochemical characteristics such as enzyme and neurotransmitter production [40, 41], which will not be discussed in this review. By combining intracellular recording and dye-injection, a direct correlation of soma size with peripheral nerve conduction velocity is possible [42]. Peripheral unmyelinated C-fibers originate from small, dark type B DRG neurons whereas peripheral Aα- and Aβ- fibers originate from large, light type A DRG neurons. Thus, type A neurons are largely proprio- and mechanosensory giving rise to A-fibers, while type B neurons are largely nociceptive and give rise to C-fibers. The average soma size of peripheral Aδ-fibers is between the average size of large, light and small, dark neuron populations, providing evidence of them being the out-of-category medium-sized neurons [42]. Trk Receptors in DRG Neurons and SGCs Distinct subpopulations of adult rat DRG neurons express TrkA, TrkB or TrkC mRNA [4, 5], although some report extendedly overlapping populations [43]. TrkA-positive neurons constitute about 40 % of DRG neurons [4, 38, 44–47] and are mostly small diameter type B neurons [38, 46, 48], as shown in Fig. 3, which depicts the DRG neuron populations

Aδ TrkC TrkA type A neurons TrkB

type B neurons

Ret Fig. 3 Expression profile of tropomyosin receptor kinase (Trk) receptors in DRG neurons. Large, light type A neurons constitute approximately 40 % of the DRG neurons (light gray area). Two distinct populations of type A neurons express TrkB and TrkC, respectively, and some TrkCpositive neurons also express TrkA. The small dark type B neurons constitute approximately 60 % of the DRG neurons (dark gray area). TrkA and glial cell line derived neurotrophic factor (GDNF) receptor Ret are expressed by distinct type B neuron populations, however, a distinct expression overlap is observed. Ret is also expressed by a small part of type A neurons. A small percentage of DRG neurons fit neither type A nor type B profiles and these neurons are likely the somas of Aδ-fibers that, at least partly, express TrkA

with respect to Trk receptor expression. A nociceptive function of the TrkA-positive population is supported by studies showing spinal cord TrkA immunoreactivity being restricted to laminae I and II [49] as well as by the abnormal pain phenotypes of NGF and TrkA knockout animals; Both homozygous NGF and TrkA knockout mice, which only survive for a few weeks postnatally, fail to respond to noxious mechanical and thermal stimuli. These mice suffer extensive loss of small and medium peptidergic neurons, i.e., type B neurons in DRGs [50, 51]. Heterozygous NGF knockout mice display a mild, albeit statistically significant phenotype, displaying a prolonged latency in the heat-sensitive tail flick test compared to wild-type mice [51]. The Aδ-fiber subpopulation of DRG neurons is suggested to be at least partially TrkA-positive as indicated by the significantly increased number of these mechanosensory Aδ-fibers in NGF-overexpressing mice [52]. Approximately 10–15 % of TrkA-positive neurons overlap with the TrkC-positive population [4]. The TrkB-positive neurons constitute 5-26 % of DRG neurons and display varied soma diameters [4, 46, 53] and are mainly belonging to the large, light type A neuron population [54]. Overall, 10–21 % of DRG neurons, almost exclusively of large size, are TrkCpositive [4, 46, 53] and are categorized as muscle afferents, i.e., proprioceptive [48, 49, 55]. P75NTR is co-expressed in most Trk receptor-positive neurons and does not seem to be expressed independently of the Trk receptors [4, 5]. A significant part of the DRG neurons does not express Trk receptors at all [5, 48]; The glial cell line derived neurotrophic factor (GDNF) receptor Ret is expressed in one third of DRG

Mol Neurobiol

neurons, constituting about half of the small, dark type B neurons [47, 56], with only a limited overlap of Ret-positive and TrkA-positive populations (reported to 13 % of the TrkApositive neurons [38]). The Ret-positive neurons project axons to the inner lamina II (IIi) of the spinal cord [57], which is specifically linked to innocuous mechanoreception [58]. However, changes of innocuous input processing in lamina IIi dorsal horn neurons have been suggested to be critical for the establishment of post-injury neuropathic pain [57, 59], and some Ret-positive neurons are designated a nociceptive modality [29]. SGCs have also been reported to express neurotrophins and their receptors. Thus, TrkA and p75NTR are found in the SGC cytoplasm and in the SGC–neuron border [60, 61], and expression of TrkB [4], more specifically truncated TrkB without the cytosolic kinase domain, has also been described [48, 53]. Furthermore, TrkC mRNA and immunoreactivity in some SGCs surrounding large neurons has also been reported [61]. Finally, NT-3, NGF and BDNF have been observed in SGCs both at mRNA and protein levels [62, 63]. Injury-Induced Changes in DRGs Profound changes can be observed in the soma of DRG neurons and in spinal cord ventral horn motor neurons upon peripheral nerve injury, ranging from altered protein expression, structural changes and even cell death depending on the degree of injury and the neuronal subtype. In general, damage to the adult PNS induces cellular mechanisms that control neuronal differentiation, growth and connectivity during development. A series of morphologic changes known as chromatolysis can be observed as early as a few hours after injury, entailing cell body and nucleolar swelling as well as nuclear eccentricity. The purpose of these changes is to initiate an appropriate response to traumatic stress, switching the neurons from the steady-state production of neurotransmitters and housekeeping proteins to a “regrowth mode”. This involves production of components for cytoskeletal and axonal reconstruction and growth-related proteins including neuropeptides, neurotrophic factors and their receptors. Also, depending on the type of injury and the distance to the soma, a considerable fraction of primary afferent neurons contributing to the injured nerve will die. Whether this is a consequence of apoptotic signaling molecules, e.g., pro-neurotrophins, lack of trophic support from target organs or a more general cellular stress resulting from the injury is yet unclear (reviewed in [43, 64–66]). Functionally, neuronal somas in DRGs often develop lowered firing thresholds or persistent spontaneous firing following injury, which is assumed to intensify peripheral signals reaching the spinal cord and, thus, may contribute to the phenomenon of sensitization and neuropathic pain [67].

The mechanisms underlying spontaneous firing are not clarified but are proposed to involve an increase in neuronal ATP sensitivity, sensitization of neighboring uninjured neurons and altered expression of transmitters. Supporting these proposals, altered communication between DRG neurons and SGCs is likely to be involved. Of the seven identified purinergic ionotropic P2X receptors (P2X1–P2X7), P2X3 is exclusively expressed by neurons and P2X7 exclusively by SGCs in sensory ganglia, which has greatly facilitated the analysis of soma-SGC communication. Thus, it is now generally believed that the neuronal soma can communicate with surrounding SGCs through bidirectional release of ATP (reviewed in [20]). In accordance with this, increased neuronal expression levels and membrane trafficking of P2X3, as well as upregulation of P2X7 expression in SGCs, have been observed following nerve injury and following NGF treatment and both are believed to be important mechanisms underlying injury-induced sensitization of DRG neurons [20, 68]. SGCs can further communicate with each other via gap junctions, which under normal physiological conditions are only observed between adjacent SGCs within a structural unit, i.e., SGCs enveloping a single neuronal soma. Following nerve injury, however, electron microscopy and injection of the fluorescent dye lucifer yellow into individual SGCs have demonstrated the formation of new gap junctions between adjacent SGCs within individual envelopes as well as gap junction bridges between SGCs in neighboring envelopes. As gap junction blockers have been found to selectively reduce spontaneous activity of DRG neurons following nerve injury, this strongly supports the importance of neuron–SGC communication on neuronal function following nerve injury [21, 69, 70]. Neurotrophin Receptors in DRGs In agreement with the concept of an injury-induced change of DRG neurons to a “regrowth mode”, numerous reports over the past couple of decades have documented dramatic changes in the expression of neurotrophins and neurotrophin receptors in sensory ganglia neurons and SGCs following injury. However, as will be described in the following sections, their responses are quite complex and reflect the type of injury paradigm and neuronal subtype but also whether the neurons suffer direct injury to their distal axon or if the distal axons are intact but affected by neighboring injured axons. Trk receptors have been found to be selectively expressed in subpopulations of sensory ganglia neurons, and axotomy has generally been reported to result in downregulation of all Trk receptors on both mRNA and protein levels in adult rodents [71, 72]. The downregulation has been found to be more pronounced in younger rats compared to older rats, which might reflect the generally higher expression levels of Trk receptors observed in younger adult rodents [71]. While expression of all Trk receptors is generally reported to be

Mol Neurobiol

downregulated in DRG neurons following injury, the situation is found to be more complex with the neurotrophins. BDNF in DRGs Several reports have demonstrated that nerve injuries greatly increase BDNF synthesis in primary afferents [56, 73–75], but it is also evident that different types of peripheral nerve injuries induce differential BDNF responses. BDNF does not appear to have an auto- or paracrine role within the DRG but is believed to be a trophic messenger that is anterogradely transported to the central terminals of primary afferents in the spinal cord dorsal horn where it may be released upon peripheral nociceptive activity inducing signal modulation and consequently pain symptoms [54, 73, 76–79]. Despite the neuronal upregulation of BDNF, only a subgroup of DRG neurons expresses BDNF prior to or as a result of nerve injury. In rats, BDNF mRNA and protein is found mainly, but not exclusively, in the subpopulation of DRG neurons that expresses the NGF receptor TrkA. TrkAnegative neurons that express BDNF include some large TrkC-positive neurons, a very limited fraction of mediumsized TrkB-positive neurons as well as a fraction of small nociceptive neurons [54]. Sciatic nerve axotomy and nerve crush have both been shown to induce a significant increase in the number of medium and large BDNF-positive neurons. However, whereas nerve crush has been found to result in both an increased number of small BDNF-positive neurons and increased BDNF intensity, nerve axotomy was found to reduce BDNF intensity in small neurons without affecting the number of BDNF-positive neurons [74]. Similarly, in a comparative study, the chronic constriction injury model on rat sciatic nerve has been observed to induce a significant increase in the percentage of small, medium and large BDNFpositive neurons in the ipsilateral L4 and L5 DRGs. On the other hand, L5 spinal nerve ligation (L5-SNL), which selectively allows injury of the L5 DRG, was shown to result in an increased number and intensity of BDNF-positive medium and large neurons in the ipsilateral L4 and L5 DRGs, but the number and intensity of small BDNF-positive neurons were found to decrease in the L5 DRG (injured), while it was found to be significantly increased in the L4 (uninjured) DRG [75, 80]. What might be the explanation for these differences? Axotomy or application of a firm ligation counteracts the communication of the proximal nerve stump with both the distal stump and the peripheral target. In contrast, both nerve crush and chronic constriction injury models allow continued contact and communication between the local neuroma and cell bodies of the injured neurons as well as with the peripheral part of the nerve and peripheral target tissue. Finally, L5-SNL induces Wallerian degeneration in the distal axons of the affected DRG neurons, which influences intact neighboring

axons and, thus, the neuronal soma in the uninjured L4 DRG. It is, therefore, likely that these aspects are causing the observed differential regulation of BDNF gene expression after cut/ligation versus crush/constriction injuries. NGF in DRGs The observation that BDNF is mainly expressed and upregulated in TrkA-positive neurons following nerve injury strongly implies the involvement of NGF as a signaling molecule in these events. This is supported by experiments in which stimulation with exogenous NGF has been found to increase BDNF mRNA and protein levels within TrkA-positive neurons, resulting in BDNF expressing in 90 % of TrkA-positive DRG neurons. As exogenous NGF has been shown not to increase BDNF expression in non-TrkA cells, it indicates that alternative signaling molecules are mediating BDNF regulation in these neurons following injury [54]. But what is the source of NGF in the DRGs? In contrast to BDNF, NGF appears to be downregulated in DRG neurons following injury [81]. However, a variety of peripheral inflammatory conditions result in locally increased NGF at both mRNA and protein levels and NGF is retrogradely transported from the injured nerve/periphery. In support of this, NGF-quenching antibodies or TrkA-Fc fusion proteins can prevent the translation of peripheral tissue inflammation into increased DRG responsiveness towards NGF [82]. The differential DRG response between cut/ligation and crush/constriction models as well as the disparity between injured and uninjured DRG responses in the L5-SNL model indicates that at least some NGF is derived from the periphery distal to the site of injury and retrogradely transported back to the DRGs via intact axons. The simplest model would be that NGF binds TrkA in the periphery to induce signaling in the TrkA-positive neurons both in the distal nerve as well as in the soma upon retrograde transport of the activated signaling complex. A competing model for NGF availability in the entire DRG might be that retrogradely transported NGF is re-released from the soma to act on neighboring neurons in the DRGs. However, such re-release is still to be demonstrated, and the tight envelope of SGCs surrounding the individual neuronal soma further argues against such a mechanism. Other evidence suggests that the periphery is not the only source of the observed NGF increase in the DRG. Following L5-SNL, a linear increase in NGF protein has been observed in the ipsilateral L4 DRG, reaching a plateau after 2 weeks. However, delayed increase in NGF mRNA in the injured L5 DRG, but not in the uninjured L4, has also been observed [75]. Combined with the observation that injured DRG neurons downregulate NGF mRNA, this suggests a non-neuronal source of NGF within the DRG upon direct injury to the distal axons. Indeed, several observations demonstrate that SGCs, but not neurons, increase NGF and NT-3 mRNA following

Mol Neurobiol

injury [63, 81, 83] and, intriguingly, administration of gap junction inhibitors during injury was found to significantly decrease the number of NGF-positive SGCs [84, 85]. In general, the SGCs respond rapidly when injury is inflicted on the neuron, which they encapsulate. Dynamic neuron–SGC communication by P2X receptors, NMDA receptors and glutamate transporters in SGCs increases SGC– SGC communication via gap junctions and initiates a general increase in proliferation and cytokine communication [9, 20, 64, 86]. This SGC response suggests that SGC involvement in post-injury events, such as regeneration and the initiation and/ or maintenance of neuropathic pain, is a highly rational assumption. In accordance with this, knockdown of the gap junction protein Connexin43 as well as the use of pharmacological gap junction blockers have been demonstrated to decrease pain behavior in rodents [3, 87–89]. The observation of SGCs also being dynamically involved in neurotrophin signaling is being increasingly recognized. Presence of TrkA, TrkB, p75NTR, NGF and BDNF in SGCs under normal physiological conditions has been described, and in situ hybridization and immunohistochemical analysis have demonstrated upregulation of NGF and NT-3 starting as early as 48 h after nerve injury and lasting at least 2 months [53, 60, 63, 69, 90]. Sympathetic Sprouting into Injured DRGs Although the direct effects of SGC-derived NGF on DRG neurons are still to be demonstrated, other observations indicate involvement of SGCs in neurotrophin signaling. Sprouting of sympathetic axons into sensory ganglia following nerve injury is believed to underlie the phenomenon of sympathetically maintained pain, where increased pain is triggered when sympathetic activity is induced. Following nerve injury, sprouts of sympathetic terminals originating from the vasculature form synaptic terminals or “baskets” around selected large DRG sensory neurons. Sympathetic nerves express and respond to neurotrophins [11, 15, 91, 92], and the observed sympathetic sprouting clearly depends on NGF as antibodies against NGF or NT-3, delivered by an osmotic mini-pump to the DRG via the transected nerve, was found to reduce sprouting significantly [81, 93]. It has been reported that uninjured and even contralateral DRGs to some extent can be affected by sympathetic sprouting [16, 81], although this might reflect that any surgical procedure per se, to some extent, may initiates local tissue inflammation and, thus, may induce systemic responsiveness to released NGF. As axotomy decreases the availability of NGF from the peripheral nerve/target, the source of NGF within the DRG is believed to be the SGCs; a model which is consistent with the observed injury-induced increase of NGF in these cells. In general, the sympathetic sprouting around the axotomized neurons is associated with p75NTR-positive SGCs [20, 21,

63]. Based on the L5-SNL model, nerve injury results in reduced p75NTR expression in injured (L5) neurons, whereas the expression increases in uninjured but affected L4 neurons. However, whereas injured neurons decrease p75NTR expression, the surrounding SGCs experience a dramatic upregulation of p75NTR levels in response to injury. Increased expression of p75NTR is primarily found in SGCs surrounding large neurons, and co-localization studies further demonstrate that sympathetic sprouting mainly associates with the p75NTRpositive SGCs surrounding these large neurons. Furthermore, other findings show that sympathetic fibers wrap up around the SGCs rather than around the neurons, and that p75NTR knockout mice display impaired sympathetic sprouting into the DRGs [23, 24, 71, 72, 81, 93, 94]. Thus, increased expression of NGF and p75NTR by SGCs potentially provides an explanation for the abnormal growth of sympathetic fibers in DRGs after peripheral nerve injury. However, whether increased NGF expression by SGCs is restricted to the p75NTR-positive SGCs surrounding large injured neurons and how SGC p75NTR is involved in the supposed SGCderived NGF attraction of sympathetic sprouts is still to be explained. Only half of the p75NTR-positive neuron–SGC units attract sympathetic sprouts, and p75NTR-positive SGCs surrounding small diameter neurons do not attract sympathetic sprouts [24, 63], suggesting that sympathetic sprouting mechanisms include, but extend beyond, SGC-related increases in NGF and p75NTR expression. Neuronal Apoptosis in DRGs One of the effects of nerve injury is a progressive loss of neurons in the affected DRGs. The consequences of nerve injury on DRG neuron numbers have been studied in both mice and rats with up to 50 % total neuron loss observed, albeit with a rather high variation, likely depending on the injury model. The neuron loss predominantly affects the type B neuron population, whereas fewer type A neurons are lost [95–102]. As many type B neurons are TrkA positive, it has been proposed that these neurons die as a consequence of reduced NGF accessibility from peripheral targets. Attempts to prevent type B neuron loss by application of local or systemic NGF has, however, been conflicting, with observed neuronal rescue in some studies [26–29, 95], but without effect in others [30, 101]. Furthermore, crush injury, which is believed to allow retrograde transport and signaling by NGF on TrkA positive neurons (see section “NGF in DRGs”), still results in a significant loss of type B neurons [101]. It has also been suggested that p75NTR is involved in the injury-induced loss of neurons [96, 101, 103, 104]. This is based on the observations that no type B neurons are lost following axotomy in absence of p75NTR utilizing either p75NTR-deficient mice or by inhibiting p75NTR mRNA translation with antisense oligonucleotides. However, the fact that p75NTR-

Mol Neurobiol

deficient mice develop serious degeneration of sensory ganglia with approximately 50 % neuron loss in the adult animal [101] might make this an inappropriate animal model to study further injury-induced neuron loss ascribed to p75NTR deficiency. A curious aspect is the finding that p75NTR is expressed in both type A and B neurons [40, 41, 105], but only type B neurons are seriously affected by injury-induced apoptosis. Furthermore, SGCs dramatically increase p75NTR expression following injury to their associated neurons, but whereas p75NTR is linked to neuronal death, SGCs proliferate following injury. It has been postulated that pro-neurotrophin-induced cell death could be the underlying cause of neuronal apoptosis, as sortilin and p75NTR are both widely expressed and are found with a high degree of co-expression in several DRG neurons [43, 105, 106]. ProNGF is also present in the DRGs [105], and the increase in neuronal BDNF synthesis following injury may provide a rich source of proBDNF in the DRGs. Only small-sized neurons co-expressing sortilin and p75NTR appear to be vulnerable to injury-induced neuronal death [38, 46, 48, 105]. Thus, the answer to the question why type B neurons are more vulnerable is currently not clear but might involve (pro)BDNF upregulation by small (TrkA positive) sortilin/ p75NTR-positive type B DRG neurons following nerve injury. Neurotrophin Signaling in DRGs Following Injury Accompanying the dynamic expression of neurotrophins and their receptors following nerve injury is a multiplicity of intracellular and cell–cell signaling in the DRGs. A general and widely used marker of neuronal injury is the induction of activating transcription factor-3 (ATF3), a member of the ATF/CREB transcription factor superfamily. After sciatic nerve axotomy in the rat, 82 % of L4 DRG neurons have been found to be ATF3-positive. It has been observed that intrathecal delivery of NGF for 2 weeks following nerve injury reduced ATF3 expression to 35 % of DRG neurons, preventing expression in a population of mainly small- to medium-sized neurons [55, 107]. This suggests that ATF3 expression may normally be induced by loss of target-derived neurotrophins. A general phenomenon following nerve injury is also the activation of the mitogen-activated protein kinase (MAPK) family of signaling molecules, which transduce various extracellular stimuli to intracellular mitogenic and differentiation signals. The MAPK family includes ERK1/2, p38, JNK and ERK5. Whereas ERK1/2 is involved in cellular growth and differentiation, p38 and JNK are involved in cellular stress such as apoptosis [9, 49]. In the L5-SNL model, axotomy was found to induce activation of ERK1/2, p38 and JNK in various populations of injured L5 DRG neurons. In contrast, only p38 activation was observed in TrkA-positive small neurons in the uninjured L4 DRG [108, 109]. Furthermore, increased phosphorylation of

TrkA was found in the uninjured L4 DRG neurons, but functional inhibition of p75NTR was found to suppress the injuryinduced phosphorylation of TrkA and p38 [4, 72]. These results demonstrate that nerve injury induces differential activation of MAPK in injured and uninjured DRG neurons, and that p75NTR might facilitate TrkA signaling in uninjured DRG neurons. Furthermore, as inhibition of p75NTR or injection of anti-NGF have been found to result in MAPK inhibition and as p38 inhibitors have been shown to suppress injury-induced neuropathic pain, MAPK activation in the DRG may participate in generating pain hypersensitivity after nerve injury [4, 46, 53, 72, 109, 110]. The increased NGF signaling in primary afferents following injury translates into potentiation of transient receptor potential cation channel subfamily V member 1 (TrpV1), an ion channel activated by heat as well as by various chemical substances such as capsaicin (the active ingredient in chili pepper) and hydrogen ions (lowered pH). In contrast to the findings that p75NTR facilitates TrkA signaling and injury-induced neuropathic pain [72, 109, 110], activation of TrpV1 by NGF appears to be mediated only by TrkA as NGF still induces TrpV1-dependent thermal hyperalgesia in p75NTR-deficient mice [54, 111–113]. However, other reports find that TrpV1 expression in small nociceptive DRG neurons is independent of TrkA expression [4, 46, 53, 114]. Similarly, the intracellular signaling cascades connecting NGF to TrpV1 activity are not clarified. Other studies have found that recruitment of PLC to TrkA is essential for NGF-mediated potentiation of TrpV1 activity whereas inhibition of protein kinase C (PKC) has been found not to have any effect [48, 49, 55, 112]. In contrast, other groups have found that inhibition of PLC does not block NGFinduced sensitization (albeit reducing the amplitude), whereas inhibition of PI3K or PKC completely abolishes the effect of NGF [4, 5, 113]. In addition, TrpV1 expression is described to be activated by the Ras/ERK pathway by some groups [5, 48, 115, 116], whereas others do not find this [47, 56, 113]. NGF is known to regulate several other pain-associated ion channels such as acid sensitive ion channels (ASICS2 and -3), voltage-gated sodium channels (Nav1.8), bradykinin receptors, endothelin receptors and various voltage-gated calcium channels (CaV 3.2, 3.3) [38, 117–119]. Thereby the present understanding of NGF involvement in hypersensitivity following nerve injury or inflammation is further complicated, prompting further investigation of the specific involvement of p75NTR and TrkA as well as intracellular signaling cascades in the DRGs.

The Peripheral Nerves Anatomy at a Glance The PNS is composed of parenchyma (nerve fibers, i.e., axons and surrounding Schwann cells) and stroma (the scaffold

Mol Neurobiol

cytoplasmic processes surrounding and segregating groups of several small-diameter axons (C-fibers with diameters of 0.15–2 μm) thereby forming the Remak bundles. Myelin covers the A-fiber axon at intervals (internodes) of 150– 1,500 μm, leaving bare gaps on the axon — the nodes of Ranvier. Schwann cells produce a basal lamina that surrounds the Schwann cell itself and the associated axon. Extracellular matrix molecules in the basal lamina, such as laminin and collagen, regulate key aspects of Schwann cell development including the formation, architecture and function of myelin [65, 123].

made of several connective elements). Nerve fibers are wrapped in the endoneurium, a loose, soft layer of delicate connective tissue made up of endoneurial cells and a collagenous matrix that embeds and protects the nerve fibers [120] (Fig. 4a). The fibers are gathered into larger groups, the fascicles, each enveloped by the perineurium, a dense and mechanically strong sheath [121]. In mammalian nerve trunks, the perineurium consists of up to 15 layers of flattened polygonal cells, collagen fibrils and capillaries and functions as a metabolically active diffusion barrier [122]. Each fascicle may contain up to several thousand axons and several fascicles are usually bundled together with blood vessels within yet another sheath, the epineurium. The epineurium is the outermost, thick layer of dense irregular connective tissue surrounding a peripheral nerve and carrying the main supply channels of the intraneural vascular system. This connective tissue contains fibroblasts, collagen and variable amounts of fatty tissue and functions to protect and support the nerve fibers (reviewed in [65]).

Wallerian Degeneration When the normal functions of the peripheral nerves are compromised by various insults such as axotomy or nerve crush, the distal segments undergo distinct morphological and molecular changes known as Wallerian degeneration [123]. The term Wallerian degeneration refers to a series of slow processes in the distal nerve stump, setting the stage for regeneration of the nerve: distal axons and myelin sheaths degenerate, Schwann cells dedifferentiate and macrophages are recruited (Fig. 4b). Within hours after nerve fiber axotomy, the axon membrane fuses and seals followed by disintegration of the distal stump within the first days. A proposed mechanism of axonal self-destruction involves nerve insults leading to increased Ca2+ influx, calcium-dependent cytoskeletal disassembly, axonal protease activation and granular disintegration [122, 124–126]. Demyelination begins with the fragmentation of compact myelin into small structures called myelin ovoids [127]. Schwann cells in adult peripheral nerves provide a

Schwann Cells Schwann cells are the main nerve-ensheathing glial cells of the PNS and have a crucial role in maintaining normal nerve function and in mediating nerve repair following injury. Schwann cells exist as two types of cells in the adult PNS, the myelinating and the non-myelinating Schwann cells, both of which enclose the neuronal axons. The myelinating Schwann cells form a multi-layered membranous myelin sheath around a segment of a single large-caliber axon (Afiber) by spirally wrapping its plasma membrane around the axon (Fig. 4a). The non-myelinating Schwann cells form

a

Nerve fascicle Perineurium Endoneurium Schwann cells

b

Injured

Proximal axon

Distal axon stump

Wallerian degeneration

Macrophages Regenerating Axonal regrowth Axon Regenerated

Epineurium

Fig. 4 Peripheral nerve structure and regeneration following injury. a Cross-section of a peripheral nerve. The epineurium encapsulates the peripheral nerve, which consists of several nerve fascicles where each fascicle is bounded by the perineurium. Several axons covered by Schwann cells and by the endoneurium constitute a nerve fascicle. b Time course of Wallerian degeneration and axonal regeneration following nerve injury. Top panel: distal to the site of neuronal injury fragmentation of the axon and myelin sheaths occurs. Second panel: during Wallerian

degeneration, recruited macrophages engulf cellular debris and Schwann cells dedifferentiate. Third panel: the injured axon begins to regrow guided by the Schwann cells, which redifferentiate into myelinating cells and encapsulate the regenerating axon. Bottom panel: under optimal circumstances, redifferentiated Schwann cells encapsulate the regenerated axon and target reinnervation is restored, allowing normal signal transmission

Mol Neurobiol

supportive milieu for axonal regeneration as they possess the ability to respond to nerve injury by dedifferentiating into their immature state and the ability to produce neurotrophic factors, two actions that facilitate successful regeneration of axons [25, 123]. Dedifferentiated Schwann cells phagocytize the generated myelin and axonal debris both independently and by recruiting circulating macrophages over a period of 3–6 weeks. In addition, Schwann cells provide a supportive cellular context for regenerative axon growth by upregulating their synthesis and secretion of neurotrophins to stimulate the regrowing axons [128–132]. During dedifferentiation, specific intracellular signaling molecules become activated and drive the dedifferentiation program: Schwann cells cease to express myelin genes, including myelin protein zero (P0) and Krox20/Egr-2, an essential transcription factor in myelination [133], and they reexpress genes found in immature states such as p75NTR [134]. Further, the transcription factor c-Jun plays a role in dedifferentiating Schwann cells not only in the demyelinating process, but also in the induction of neurotrophic factors such as GDNF and Artemin [135]. An essential process is the activation and recruitment of macrophages, which are involved in the clearance of axonal and myelin debris by phagocytosis. Activation of Toll-like receptors on Schwann cells is needed upon nerve injury to stimulate Schwann cells to produce proinflammatory mediators including TNFα, IL-1β and IL-6, which are important for recruitment of immune cells such as macrophages [136, 137]. Furthermore, secretion of metalloproteases and vasoactive mediators by resident macrophages and injured axons causes breakdown of the blood–nerve barrier, and circulating macrophages are recruited from the vasculature into the site of injury [137, 138]. Peripheral Nerve Regeneration Peripheral nerve regeneration requires activation of the intrinsic growth capacity of the injured neurons in order for axonal regrowth and target re-innervation to occur. Furthermore, remyelination of the regrowing axons is important for successful functional recovery after nerve injury. Following Wallerian degeneration, dedifferentiated Schwann cells proliferate and migrate, aligning in Bands of Büngner (Schwann cell tubes) within the basal lamina of the denervated distal endoneurial tubes. The Bands of Büngner are crucial for axonal regeneration as they provide trophic support, including secreted neurotrophins, for the regenerating axons and secrete extracellular matrix molecules like laminin and collagen as well as cell adhesion molecules (reviewed in [139]). Regrowing axons elongate from the intact proximal part of the nerve trunk across the injury site and into the Bands of Büngner. Consequently, axonal regeneration is severely compromised following axotomy, which involves damage to

the endoneurium [140, 141]. In contrast to axotomy, a crush injury usually leaves the endoneurium intact and axons can, therefore, regenerate in their native endoneurium, leading to improved regeneration and target reinnervation. The dedifferentiated Schwann cells redifferentiate upon contact with the regenerating axons, ensheath and subsequently remyelinate the axons or restore Remak bundles thereby facilitating functional recovery [139]. Neurotrophins are crucial in relation to axonal regrowth and remyelination, and their specific roles in these processes are beginning to be unveiled. Neurotrophins and Axonal Regrowth The spatial and temporal expression of neurotrophins and their receptors has been examined in the injured nerves and the main focus has been on elucidating their importance in relation to regeneration of the sciatic nerve and the median nerve, which both contain sensory and motor axons. Sensory and motor neurons have different requirements for neurotrophic support in intact as well as injured states, and techniques such as retrograde labeling allow separate analysis of neurotrophin requirements for regeneration of both axon types. As described in the section “Trk Receptors in DRG Neurons and SGCs”, sensory neurons can be divided into subpopulations depending on their need for neurotrophins. Accordingly, nociceptive and thermoceptive DRG neurons require NGF for survival [51], whereas proprioceptive neurons require NT-3 [142, 143]. BDNF and NT-4/5 are potent survival and trophic factors for intact as well as injured motor neurons in vivo [144–147]. BDNF in Axonal Regrowth Following nerve injury, a continuous slow monophasic increase of BDNF mRNA is observed in the distal segment of the rat sciatic nerve starting 3 days after axotomy and reaching maximal levels after 2–4 weeks [130, 148]. Several studies suggest that Schwann cells of the distal nerve stump synthesize BDNF and that they are responsible for the BDNF increase following injury [130, 148–150]. However, DRG neurons reportedly transport BDNF anterogradely in both peripheral and central processes [151], thus, potentially contributing to the BDNF increase as anterograde transport of BDNF is significantly enhanced after peripheral nerve injury [152]. BDNF-deficient mice die shortly after birth [153]. Thus, to examine the role of BDNF in axonal regrowth following peripheral nerve injury, research has been constrained to employ heterozygous BDNF mice and pharmacological manipulation of BDNF. The role of endogenous BDNF in axon regrowth from the sciatic nerve has been examined by depriving endogenous BDNF with BDNF-neutralizing antibodies. This treatment has been shown to result in reduced elongation of regenerating axons following sciatic nerve crush injury,

Mol Neurobiol

which indicates that endogenous BDNF is important for stimulating axonal elongation [149]. Accordingly, axonal regeneration can be enhanced by repairing transected nerves of the common fibular nerve with nerve grafts to which exogenous BDNF has been applied [154]. The growth of axons through a common fibular nerve graft from a heterozygous BDNF knockout mouse is not significantly affected; however, it indicates that even reduced BDNF levels are sufficient to promote normal axonal growth [154]. These studies do not discriminate between sensory and motor neuron axonal regrowth when evaluating the role of BDNF. One approach to specifically study motor neuron axonal regeneration in mixed nerves is by applying retrograde tracers to transected nerve fibers and subsequently quantifying labeled motor neuron somas in the corresponding spinal cord segment. This approach has been employed to examine the role of various doses of exogenously applied BDNF on motor neuron axonal regrowth after transection of the tibial nerve. Low doses of exogenous BDNF has been shown to promote motor neuron axonal regeneration following repair of longterm transected nerves [155] whose capacity to regenerate is generally poor [140, 156]. In contrast, high doses of BDNF have been shown to significantly inhibit motor neuron axonal regrowth following nerve repair both immediately and after long-term transection. Thus, BDNF is suggested to exert dosedependent facilitation and inhibition on motor neuron axonal regrowth. The BDNF receptor TrkB appears to be critical for motor neuron axonal regeneration as axonal regrowth is significantly attenuated in heterozygous TrkB mutant mice. In contrast, function-blocking antibodies against p75NTR have been reported to increase motor neuron axonal regeneration and to reverse the inhibitory effect of BDNF, indicating an inhibitory role of p75NTR on motor neuron axonal regrowth [155]. Mature motor neurons express only low levels of p75NTR; however, expression is upregulated in regenerating motor neurons after peripheral nerve injury [157, 158]. Expression of p75NTR is triggered by retrograde transport of positive signals derived from axons regrowing through damaged or denervated peripheral nerve tissue [159], and it is downregulated when motor function is restored [158]. Similarly, p75NTR mRNA increases markedly in Schwann cells of the distal segment of the transected sciatic nerve, and the expression of p75NTR is downregulated to pre-injury levels when regenerating axons make contact with the Schwann cells of the distal nerve trunk [160, 161]. P75NTR expression in Schwann cells appears to be important for motor neuron axonal regrowth as a lower number of retrogradely labeled motor neurons is observed after sciatic nerve repair with a nerve-graft containing Schwann cells deficient of p75NTR [162]. The role of Schwann cell-derived BDNF in regeneration of motor neuron axons has recently been examined by analyzing regeneration of wild-type motor neuron axons through grafts

of BDNF-deficient Schwann cells. The length of regenerating axons into grafts of BDNF-deficient Schwann cells have been found to be significantly shorter than those growing into wildtype grafts. Addition of recombinant human BDNF at the site of repair were shown to rescue axon regeneration in absence of Schwann cell-derived BDNF as well as to enhance the length of regenerating axons into wild-type grafts [163], indicating that BDNF derived from Schwann cells is important for motor neuron axonal regeneration after nerve transection. It has been suggested that the enhancement of axonal regeneration is mediated via retrograde signaling of Schwann cell-derived BDNF acting on TrkB receptors on the growing axons [163]. In line with this hypothesis, Schwann cells express truncated non-catalytic TrkB receptors [147] and are suggested to bind and present BDNF to motor neuron axons. As described in the section “BDNF in DRGs”, BDNF is expressed by uninjured sensory neurons in the SNL model, and endogenous BDNF has, thus, been hypothesized to be responsible for activating the intrinsic growth capacity of DRG neurons following peripheral nerve injury. By neutralizing endogenous BDNF with BDNF siRNA or BDNF antibodies immediately prior to and following spinal nerve injury, respectively, a reduced injury/regeneration-associated gene expression and a reduced intrinsic capacity of these neurons to extend neurites in vitro have been detected. Endogenous BDNF is, however, not critical for maintaining the soma response to injury, as delayed infusion of BDNF antibody does not alter injury/regeneration-associated gene expression once it has been initiated. These results suggest BDNF as being critical for the initial induction of the regenerative response in injured sensory neurons [164]. Furthermore, BDNF enhances neurite outgrowth of DRG neurons in vitro, whereas exogenous and endogenous proBDNF collapses neurite outgrowth by activating the small GTPase RhoA and its downstream effector Rho kinase (ROCK) via p75NTR [165]. NGF, NT-3 and NT-4/5 in Axonal Regrowth The role of NGF, NT-3 and NT-4/5 with regard to axonal regrowth following peripheral nerve injury has, to the best of our knowledge, been less extensively investigated as compared to BDNF. NGF mRNA and protein are not detected in the intact adult rat sciatic nerve but expression is markedly induced in non-neuronal cells of the distal and the immediate proximal segment of transected nerves [131, 166]. The levels of NGF mRNA exhibit a biphasic response to axotomy with an immediate early rise in NGF mRNA peaking 6 h after injury. The second increase in NGF mRNA expression is slower and is believed to be caused by IL-1, released by macrophages invading the site of injury during Wallerian degeneration and lasting several weeks [131, 167]. Studies

Mol Neurobiol

indicate Schwann cells as being the source of NGF [130, 148, 166, 167] in the transected nerve. The induction of p75NTR expression in Schwann cells distal to the site of axotomy [160, 161] has been proposed as a mechanism for Schwann cells to act as substitute targets for NGF-dependent neurons by concentrating and presenting NGF to regenerating NGFresponsive TrkA-positive sensory neurons [161, 168]. However, this hypothesis has been challenged by a study showing that anti-NGF treatment of crushed nociceptive axons does not affect axon regrowth or functional recovery [169]. The NT-3 level in nerve tissue has been shown to decrease shortly after nerve transection, but to return to the normal level 2 weeks post injury [130, 148]. As is the case for NGF, studies indicate Schwann cells as being the source of NT-3 in the transected nerve [170, 171]. Exogenously applied NT-3 has been found to prevent reduction of the conduction velocity of axotomized myelinated sensory neurons. Thus, NT-3 appears to promote regeneration of sensory neurons [84]. Moreover, NT-3 is important for survival of denervated Schwann cells in the distal segment of the injured nerve. Mature Schwann cells in adult nerves are able to survive in absence of axons by establishing an autocrine survival loop [170]. NT-3 is one of the important factors of the autocrine regulatory mechanism that support long-term Schwann cell survival [170, 171], nerve regeneration and remyelination after peripheral nerve injury [171]. However, prolonged denervation results in Schwann cell atrophy and, thus, a decreased axonal regeneration capacity due to loss of receptors and reduced expression of growth factors by Schwann cells [141, 172, 173]. Peripheral nerve injury has been reporter to result in downregulation of the NT-4/5 mRNA level shortly after injury. However, 2 weeks post injury the NT-4/5 mRNA level has been found to be upregulated in the distal nerve stump of the injured rodent sciatic nerve [130, 174]. Studies performed using grafts from homozygous or heterozygous NT-4/5 knockout mice incorporated into wild-type sciatic nerves have shown that even a small reduction in endogenous NT-4/5 results in a significant reduction of regenerating motor neuron axonal growth [154]. In line with these results, application of NT-4/5 to heterozygous NT-4/5 knockout mice at the time of surgical repair of a cut nerve has been found to restore motor axon growth [154]. Moreover, application of NT-4/5 to repaired rat sciatic nerves has been reported to result in enhanced axon regeneration observed by an increase in the number of regenerated axons and to improve axonal diameter and myelin thickness [175]. Schwann Cell Phenotype and Modality-Specific Regeneration Traditionally, Schwann cells are described as being either myelinating or non-myelinating [176]. However, studies indicate that Schwann cells express distinct sensory and motor

phenotypes according to growth factor expression and their ability to stimulate sensory and motor neuron axonal regrowth. Schwann cells of cutaneous nerves (containing axons of sensory neurons) and Schwann cells of ventral roots (containing axons of motor neurons) differ in growth factor profiles in both intact and injured nerves. For example, NGF and BDNF are upregulated in cutaneous nerves but minimally in ventral roots after nerve injury [177]. Accordingly, Schwann cells of sensory and motor phenotypes preferentially support cutaneous and ventral root axonal regeneration, respectively [177]. Recent evidence suggests that the Schwann cell phenotype varies even more as the profiles of growth factor expression by denervated Schwann cells have been found to vary in magnitude and pattern dependent on central or peripheral location and on the modality of their associated axons [178]. Neurotrophins and Myelination of Peripheral Nerves In addition to regrowth of axons, successful regeneration of injured peripheral nerves requires remyelination. The function of myelin is to maximize the efficiency and velocity of action potentials along the distance of axons, the importance of which becomes apparent from demyelination diseases such as multiple sclerosis and Charcot–Marie–Tooth disease [179, 180]. Peripheral myelin is formed by Schwann cells that wrap around regrowing axons in a complex, dynamic process that involves neuron–Schwann cell crosstalk. The process of peripheral myelination can be separated into three major phases of Schwann cell growth and differentiation. The first phase, the proliferative stage, is characterized by proliferation and migration of Schwann cells on axons. In the second phase, the premyelination stage, Schwann cells elongate and ensheath the axons. The third and final phase, the myelination stage, is characterized by formation and growth of the myelin sheath [181]. Neurotrophins are important for both migration of Schwann cells and for Schwann cell myelination during development and for regeneration of nerves following peripheral nerve injury. Neurotrophins and Schwann Cell Migration — the Proliferative Stage Schwann cells express, in addition to p75NTR [160, 161], TrkC and a truncated non-catalytic form of the TrkB receptor. The levels of Trk receptors are higher in the distal part of the transected sciatic nerve compared to the proximal part of the injured nerve [130]. BDNF and NT-3 are implicated in Schwann cell migration in co-cultures of Schwann cells and DRG neurons as the migration of Schwann cells is significantly enhanced by NT3 through activation of TrkC [182] and inhibited by BDNF signaling through p75NTR [183]. The differential effects of

Mol Neurobiol

BDNF and NT-3 on Schwann cell migration are regulated through signaling pathways that depend on Rho GTPases. Specifically, NT-3 activation of TrkC enhances the migration of primary Schwann cells through the signaling pathway regulated by the Rho GTPases Rac1 and Cdc42. Endogenous BDNF acting through p75NTR inhibits Schwann cell migration dramatically by Src kinase-dependent activation of the guanine-nucleotide exchange factor Vav2 and RhoA [183]. In addition to NT-3, NGF also appears to facilitate Schwann cell migration, although in a p75NTR-dependent manner [184]. In line with these results, p75NTR is important for regulating Schwann cell migration during development of peripheral nerves [185]. Neurotrophins and Myelination — Developmental and In Vitro Studies Neurotrophins have been identified as important regulators of Schwann cell myelination both in vitro and in vivo during peripheral nerve development. NGF has been identified as a potent regulator of axonal signals that control myelination of TrkA-expressing DRG neuron subpopulations in vitro, thereby promoting myelination by Schwann cells [186]. The roles of BDNF and NT-3 on myelination have been examined in Schwann cell/DRG neuronal co-cultures. NT-3 levels are initially high, but are downregulated throughout the proliferation and the premyelination stages, whereas BDNF levels correlate with myelin formation. Moreover, exogenously applied BDNF enhances myelination, whereas application of NT-3 to the co-cultures inhibits myelination. In line with these results, removal of endogenous BDNF has been shown to inhibit myelination, whereas removal of endogenous NT-3 has been shown to increase myelination. Similar results have been observed in vivo in the developing mouse sciatic nerve [187]. Accordingly, adenoviral-mediated overexpression of BDNF in DRG neurons have been reported to enhance myelination in Schwann cell/DRG neuron co-cultures [188]. These results suggest that BDNF is a positive modulator of myelination and that NT-3 is a negative modulator of myelination [187]. The myelination-promoting effect of BDNF is mediated by p75NTR, whereas NT-3 mediates its myelination-inhibitory effect via TrkC. A truncated isoform of TrkB is upregulated during the myelination stage and acts as an inhibitory regulator of myelination by competing with p75NTR for the availability of endogenous BDNF. In agreement with these results, myelination is reduced upon treatment with an inhibitor of p75NTR and in homozygous p75NTR knockout mice [189]. Neurotrophins and Myelination — the Myelination Stage Considering the expression patterns of neurotrophins and neurotrophin receptors following peripheral nerve injury as

well as their roles in in vitro myelination and myelination during development, it is highly plausible that these factors play similar roles during remyelination of regenerating peripheral nerves. Especially BDNF and p75NTR have been examined and appear to be important players in remyelination after peripheral nerve injury. Endogenous BDNF can be neutralized by treatment with BDNF antibodies, resulting in an approximately 80 % reduction of the number and density of myelinated axons in the distal part of the injured sciatic nerve [149]. Likewise, p75NTR is important for remyelination after injury. Sciatic nerve crush experiments on p75NTR knockout mice have been found to result in a reduced number of myelinated axons, in reduced myelin sheath thickness as well as in reduced levels of myelin gene expression in regenerated nerves [190]. In particular, p75NTR expression in Schwann cells appears to be important for remyelination as poor myelination has been observed when p75NTR-deficient Schwann cells were grafted onto wild-type sciatic nerves [162]. Therapeutic Approaches Enhancing Peripheral Nerve Regeneration The PNS has significant regenerative potential allowing recovery after injury, although with varying degrees of success depending on type and severity of the injury. Peripheral nerve injury leads to a rapid and robust increase in neurotrophin synthesis in neurons and Schwann cells, which guide and support regenerating axons. Efforts are being made to take advantage of this to improve nerve regeneration. Various approaches of neurotrophin application have signified the important role of the neurotrophins during the early stages of regeneration and support the significance of developing combined gene and cell therapy for peripheral nerve repair. NGF — Therapeutic Approaches The expression of NGF in transected peripheral nerves increases in the distal and immediate proximal part within the first few weeks after injury [131], implying that initial basal levels of NGF could be a limiting factor, and that supplementation of NGF could have a beneficial effect on regeneration. Several experimental models have been used to investigate the effects of NGF on peripheral nerve regeneration in rodents, including direct NGF application [191], NGFcontaining fibrin sealants [192], injection of NGF-encoding lentiviral [191–194] or adenoviral [195] particles, microspheres [196] and modified scaffolds [197, 198]. These studies have shown positive effects on regeneration, but often with conflicting effects on both sensory and motor neurons. An alternative may be Schwann cell-based therapy as engineering Schwann cells for increased neurotrophin production facilitates their regenerative effect and renders them a potent tool to guide and support nerve regeneration. To

Mol Neurobiol

optimize neurotrophin supply in the early stages of regeneration, a cell-based NGF gene therapy model has recently been tested in which Schwann cells robustly and continuously overexpressing NGF were transplanted onto the rat sciatic nerve upon transection. The transplanted Schwann cell were found to lead to significantly increased NGF levels and to a substantial improvement in axonal regeneration of a sub-class of sensory neuron populations resulting in reduced denervated muscle atrophy [199]. NT-3 — Therapeutic Approaches The ability of NT-3 to reverse axotomy-induced changes in adult motor and sensory neurons has been examined using the physiological measure of conduction velocity. Five weeks after axotomy, sensory and motor neuron conduction velocities have been found to be greatly reduced, however, application of NT-3 directly onto the transected nerve stump has been shown to largely prevent this and further to rescue motor neurons. This amelioration of physiological deficits in adult mammalian neurons suggests possible therapeutic applications of NT-3 [84]. NT-3 is an important component of the autocrine survival loop supporting Schwann cell survival and differentiation in absence of axons. Nerve regeneration deficiencies have been identified in NT-3 heterozygous knockout mice characterized by fewer Schwann cells in the regenerating nerve fibers of crushed sciatic nerves [171]. NT-3 heterozygous knockout mice display retardation of the myelination process, and this defect has been found to be associated with decreased Schwann cell survival and increased neurofilament packing density of regenerating axons. These observations indicate that the NT-3 status of the Schwann cells, but not of the axons, is responsible for impaired nerve regeneration and that NT-3 is essential for Schwann cell survival in early stages of regeneration-associated myelination in the adult rodent peripheral nerve [200]. Consistent with this hypothesis, the impact on axonal regeneration of the rat peroneal nerve after injury has been investigated using genetically modified peripheral nerve grafts repopulated ex vivo with Schwann cells modified to express NT-3, which has been found to result in a significant increase in the number of sensory fibers [201]. It has been demonstrated that xenograft transplants of Schwann cells from patients with Charcot–Marie–Tooth disease type 1A (CMT1A) into the sciatic nerve of nude mice delays myelination and impairs regeneration of axons passing through the grafted segments. The efficacy of NT-3 treatment using this model was evaluated by the number of myelinated fibers and Schwann cells, and NT-3 treatment was found to augment axonal regeneration. Furthermore, the effect of NT-3 on regeneration has been tested in patients with CMT1A by subcutaneous administration of NT-3. In this pilot clinical trial, it was shown that patients may benefit from NT-3

treatment, as regeneration in sural nerve biopsies before and after treatment was reported to increase the mean number of small myelinated fibers [171]. NT-4/5 and BDNF — Therapeutic Approaches The growth of regenerating axons is very poor if the surrounding Schwann cells are devoid of one or more of the neurotrophins or their receptors [155, 202] or if the Schwann cells are destroyed [203–205]. In line with this, topical application of either recombinant human BDNF or NT-4/5 to allografts from knockout mice at the time of surgical repair has been shown to overcome these deficits [154]. Similarly, electrical stimulation resulting in increased expression of BDNF and TrkB in motor neurons [206] and DRG neurons [203, 207] has been reported to result in enhancement of axonal regeneration following peripheral nerve transection [206, 208]. Furthermore, the regenerating axons have been found to grow more than twice as far during the first 2 postrepair weeks if the proximal stump of a cut nerve has been stimulated for merely 1 h at the time of surgical repair [203]. Finally, modest treadmill training has been reported to have a positive effect on axon regeneration [209] with a magnitude comparable to that obtained by electrical stimulation [210]. NT-4/5 in the proximal stump of the cut nerve is implicated in the treadmill training-induced enhancement of axon regeneration, while regeneration is independent of NT-4/5 presence in the tissue through which the axons grow [211]. Following peripheral axotomy, Schwann cells in the nerve segment distal to the injury synthesize and secrete BDNF into the Schwann cell basal lamina [130]. Initially, this Schwann cellderived BDNF is used by the regenerating axons entering endoneurial tubes in the distal segment to initiate axonal elongation. Spinal cord motor neurons contain BDNF and TrkB mRNAs [206, 212] with initially increased expression of BDNF following axotomy but decreasing again within 1 week [65, 130]. Schwann cells in the distal stump, however, can synthesize and secrete BDNF for 2 months or longer, and since dedifferentiated Schwann cells express the truncated non-catalytic TrkB receptor, stimulation of axon elongation is proposed to occur via a classic retrograde signaling [163, 213, 214]. In support of this, two smallmolecule TrkB agonists, 7,8-dihydroxyflavone [215] and deoxygedunin [216], have recently been found to enhance axon regeneration and to improved muscle reinnervation following peripheral nerve transection [217].

The spinal cord Anatomy at a Glance The murine spinal cord extends from the hindbrain at the foramen magnum to the lower part of the vertebral column

Mol Neurobiol

ending at the level of the sixth lumbar vertebra [218]. Thus, the spinal cord does not reach the end of the vertebral canal, and the spinal nerves form the cauda equina in the caudalmost region of the vertebral canal. The general organization of the vertebral column is conserved among mammals, but interstrain and inter-species variations exist. In the light of the use of various mouse strains and rats for research, this may be important depending on experimental setup [18, 219–223]. As the spinal cord is shorter than the vertebral canal, only the rostral-most spinal segments correspond to the vertebrae numbers, whereas more caudally spinal cord segments are located more rostrally than the vertebrae numbers indicate. This discrepancy increases in rostro-caudal progression and increases from birth to maturity [224]. The spinal cord forms two clearly visible enlargements: the cervical enlargement (generally C5– T1) and the lumbar enlargement (generally L2–L6), which consist of the segments in which the nerves of the limbs connect to the spinal cord. The spinal cord is covered by three spinal meninges: the dura mater (outermost layer), the arachnoid mater (intermediate layer) and the pia mater (innermost layer). The subarachnoid space containing the cerebrospinal fluid is located between the arachnoid mater and the pia mater, while the epidural space is located between the dura mater and the periosteum of the vertebral canal. On each lateral side of the rodent spinal cord, the dorsal and ventral rootlets are attached and fuse to form the dorsal and ventral roots, respectively [225]. Each dorsal root enters a DRG, although dorsal rootlets in the cervical region may directly enter the DRG without forming a dorsal root [225, 226]. Each ventral root fuses with its corresponding dorsal root slightly distal to the DRG (Fig. 1). The dorsal roots consist of primary afferent sensory fibers and the ventral roots of efferent somatic motor axons [226]. The mature spinal dorsal horn can be divided into laminae I–V [227–229] and it is widely recognized that small, unmyelinated and thinly myelinated neurons, i.e., Aδ- and C-fibers that transmit pain and temperature, mainly terminate in the superficial laminae (I–II), while Aα- and Aβ-fibers that transmit proprioceptive information as well as innocuous touch and pressure mainly terminate in deeper laminae [230]. The connectivity in the spinal cord is rather complex with numerous inhibitory or excitatory interneurons modifying the afferent signals through GABAergic/glycinergic inhibition or glutamatergic excitation, respectively (reviewed in [231]). Hence, the final signal to the brain is a delicate balance between stimuli from the periphery modulated by excitatory and inhibitory signals in the spinal cord. Spinal Cord Neurotrophins and Neurotrophin Receptors P75NTR is expressed in spinal cord motor neurons, and following peripheral nerve injury, p75NTR expression increases

in their cell body as well as in their axons [232]. In addition, p75NTR is expressed in oligodendrocytes where it can induce cell death following stimulation by NGF [233, 234]. TrkA is in particular observed in the superficial dorsal horns (laminae I and II), but small amounts are also found in distinct areas of deeper laminae extending to laminae IVand V depending on the location along the spinal cord [49, 235]. TrkA expression is confined to a small subset of myelinated neurons, hence, TrkA and non-TrkA expressing neurons may represent functionally distinct nociceptor populations [49]. In addition, TrkA expressing neurons have been found to coexpress p75NTR [235]. NGF is expressed only at relatively low levels in the normal spinal cord but NGF transcript levels are upregulated as a response to peripheral nerve injury [158]. In addition to signaling via TrkA, NGF has been suggested to be important for spinal purinergic signaling in that intrathecal NGF administration has been shown to increase expression of purinergic receptor P2X3 on laminae I and II nociceptors as well as immediately ventrally to the central canal [236]. Hence, NGF may contribute to chronic pain development following peripheral nerve injury. In the normal spinal cord, BDNF transcripts have been found in lamina IX α-motorneurons and to a lower extent throughout the gray matter [237]. Its cognate Trk receptor, TrkB, is expressed on lamina I neurons but also in deeper laminae of the spinal cord [238], and TrkB transcripts are also observed in motor neurons [239]. Upon peripheral nerve injury, TrkB levels have been shown to be upregulated in both neurons and activated microglia in the spinal cord dorsal horn [78, 240]. BDNF is well known to be involved in modulation of pain processing in the CNS [241, 242] (reviewed in [82, 243]) and TrkB signaling is important for neuropathic pain development induced by peripheral nerve injury and inflammation [244, 245]. In line with this, abrogation of TrkB functioning has been shown to attenuate thermal and mechanical hypersensitivity [244, 245]. Following noxious stimuli, not only TrkB levels but also BDNF levels increase in the spinal cord dorsal horn [80, 246, 247] leading to phosphorylation of the downstream protein ERK1/2 in spinal cord dorsal horn neurons and the spinalothamic tract [78, 248]. Neurons have been shown to secrete BDNF [249] and activated microglia secrete both proBDNF and BDNF [250, 251]. Several alternatively spliced TrkB isoforms exist [252], where TrkB.T1 is the predominant isoform found in the adult mammalian nervous system. TrkB.T1 differs from the fulllength isoform by lacking the intracellular kinase domain, but extracellular high-affinity BDNF binding can still occur [252, 253]. Hetero-dimerization of TrkB.T1 with the full-length TrkB isoform inhibits TrkB autophosphorylation necessary for downstream signaling. Hence, it has been suggested that TrkB.T1 may function to reduce BDNF signaling [254–256] while others have suggested that TrkB.T1 may signal

Mol Neurobiol

independently [257–260]. A study on inflammation has shown TrkB.T1 upregulation in the spinal cord dorsal horn [261] and in vitro studies on brain glial cells have revealed truncated TrkB receptor transcripts in astrocytes and oligodendrocytes but not in microglia [213]. A study on brain astrocytes has found expression of TrkB.T1 upon BDNF stimulation to be involved in calcium signaling [262]. Hence, truncated TrkB may participate in and possibly modify nociception. Transcripts of the other TrkB ligand, NT-4/5, are robustly observed in spinal cord dorsal horn laminae III– VII, to a lower extent in superficial laminae I and II and densely throughout the ventral horn of rat lumbosacral spinal cord. Furthermore, NT-4/5 transcripts are found in both neurons and in subpopulations of glial cells [237]. TrkC and its high-affinity ligand NT-3 are also found in the spinal cord [237, 239, 263]. TrkC expression has been found in spinal motor neurons [239], and following peripheral nerve injury TrkC transcript levels have been found to increase [130]. In normal adult rats, NT-3 mRNA expression has been observed to be restricted to the spinal cord gray matter in a similar pattern to BDNF expression [237] in that transcripts have been found most densely within lamina IX α-motorneurons and at lower levels elsewhere in the gray matter. Furthermore, NT-3 mRNA is also expressed by spinal cord glial cells but to a lesser extent compared to NT-4/5 mRNA [237].

Central Sensitization Following Peripheral Nerve Injury Primary afferents transmit signals from their peripheral sensory target tissue to the spinal cord dorsal horn [264]. Various changes in the spinal cord may contribute to spinal cord pathology following peripheral nerve injury, known as central sensitization. Such changes are believed to result from disinhibition, possibly caused by death of inhibitory interneurons in the dorsal horn following injury [265, 266] or by dysfunctional inhibitory signaling (reviewed in [267]) ultimately leading to the perception of non-noxious stimuli as being painful. Inflammatory mediators such as cytokines play a critical role in cell communication following nerve injury, and neuron– glia interactions are believed to be critical for establishing and maintaining injury-induced hypersensitivity. Microgliosis (outlined in the following section) induced by peripheral nerve injury is orchestrated through signals released by neurons as well as by astrocytes and immune cells. It appears that signals from injured neurons as well as from uninjured neighboring sensory neurons that share the same nerve trunk are critical for microgliosis development [268, 269], and signaling molecules including growth factors, a variety of cytokines, ATP, NO, complement components and reactive oxygen species facilitate direct neuron–microglia crosstalk (reviewed in [270]).

Microgliosis and Astrogliosis Spinal cord resident glial cells, i.e., microglia and astrocytes, play substantial roles in spinal cord pathogenesis as a response to peripheral nerve injury, ultimately affecting GABAergic and glycinergic inhibition and contributing to neuronal disinhibition. It has been demonstrated by injection of activated microglia into the spinal cord which was found to be sufficient to induce neuronal hypersensitivity [271]. Microglia constitute approximately 10 % of the total number of cells in the CNS and are considered CNS-resident immune cells originating from macrophages of the embryonal yolk sac [272]. Microglia can be divided into various groups according to their activation states: from the surveillance state to the “activated” response state with amoeboid phagocytic function. In the surveillance state, their morphological structure displays fine and long processes and a small soma. In case of altered homeostasis of the CNS, e.g., elicited by a peripheral nerve injury, they rapidly respond by changing their morphology to be amoeboid-like with retracted processes, changing their protein expression profile and increasing in number by proliferation and migration — collectively known as microgliosis [270, 273–275]. Numerous cells and signaling molecules are involved in microgliosis, including a variety of cytokines and the cytokine subfamily of chemokines (reviewed by [276, 277]). Other studies have shown that stimulation of microglial sortilin by neurotensin promoted microglial migration and their ability to secrete cytokines [278, 279]. Peripheral nerve injury triggers microgliosis that can be observed in the spinal cord dorsal horn as well as in the spinal cord ventral horn around the cell bodies of injured motor neurons ipsilateral to the injury [280, 281] as early as 24 h following nerve injury and being well established by day 3 post-injury [282] (Fig. 5). Microgliosis is confined to the spinal cord segments where injured primary afferent neurons fuse with the spinal cord [283]. Upon peripheral nerve injury, expression of a microglial activation marker, the purinergic P2X4 receptor, increases [271]. P2X4 receptor activation has been shown to be involved in mechanical allodynia development [284] by ATP-induced microglial BDNF release [251], albeit the origin of P2X4 receptor activating ATP is not clear [285, 286]. Not only BDNF but also proBDNF has been shown to be released by activated microglia [250, 251]. Astrocytes also become activated following injury [287] although at a slightly later time point than microglia [288, 289]. Astrocytes are the most numerous cells among CNS glial cells [290] and are essential for the maintenance of a functional environment in the CNS [291, 292]. Like microglia, astrocytes are able to change morphology and become more dense in response to neuronal injury, metabolic insults or inflammation [292]. Activated microglia release IL-18 that acts on astrocytes and plays an important role in generation

Mol Neurobiol

a

Cl-

Lamina I neuron

Cl-

Cl-

Cl-

Cl-

ClCl-

GABA(A) receptor

KCC2 TrkB

Cl-

Cl-

Cl-

KCC2

Low intracellular chloride concentration

b

Cl-

Activated microglia

Cl-

Cl-

BDNF

Cl-

Cl-

KCC2

Cl-

Cl-

GABA(A) receptor

Cl-

TrkB dimerization Cl-

Cl-

Cl-

Cl-

High intracellular chloride concentration

Cl-

Cl-

Fig. 5 a In the normal spinal cord, GABA(A) receptors mediate neuronal chloride influx while the potassium–chloride co-transporters KCC2 mediate chloride efflux. This maintains intracellular chloride concentrations below the equilibrium potential for chloride, i.e., neuronal hyperpolarization is achieved. b Upon peripheral nerve injury, microgliosis is

initiated (left). Activated microglia secrete BDNF, which acts on TrkB to initiate incompletely understood pathways leading to significantly decreased KCC2 levels (right). Consequently, intracellular chloride concentrations rise and GABA-mediated neuronal inhibition is attenuated, ultimately contributing to neuropathic pain development

of tactile allodynia [293]. Hence, microglia activation contributes to astrocyte activation, and continuous crosstalk between microglia and astrocytes involved in maintenance of spinal cord pathology following nerve injury is generally observed [290].

sortilin in the spinal cord [301] indicates a potential role of proBDNF in spinal cord signaling upon peripheral nerve injury. Under uninjured conditions, the neuron-restricted potassium–chloride co-transporter KCC2 establishes a chloride gradient by maintaining the intracellular chloride concentration below the equilibrium potential for chloride. Upon activation by GABA, the GABA(A) receptors mediate influx of chloride ions down their electrochemical gradient. This renders the neuronal membrane potential more negative relative to the resting membrane potential, i.e., hyperpolarization is achieved, and the chance of generating a successful action potential is diminished. Hence, KCC2 is critical for establishment of the chloride gradient that is needed for GABAergic and glycinergic inhibition in adult neurons [302, 303]. In the spinal cord, glycine receptors and GABA(A) receptors colocalize at postsynaptic densities [304], display similar chloride and HCO3− permeability [305] and both mediate inhibitory neurotransmission [306, 307]. Therefore, glycine receptors are believed to contribute to intracellular chloride concentrations in a similar manner to GABA(A) receptors both in normal conditions but also following nerve injury [302]. Upon nerve injury, microglia-derived BDNF binds to TrkB at the neuronal cell surface resulting in TrkB dimerization and

Effects of Spinal Cord BDNF Following Peripheral Nerve Injury Microglial BDNF released following peripheral nerve injury has been found to depend on microglial p38 MAPK activation [294–296]. In addition, in vitro studies suggest a role for the adenosine A2A receptor in controlling BDNF release from activated microglia [297]. To the best of our knowledge, no reports of proBDNF involved in spinal cord signaling following peripheral nerve injury have yet been made in relation to neuropathic pain, and it is BDNF that has been found at terminals of primary afferents in the superficial dorsal horn and to a lesser extent in deeper laminae [298]. However, in the hippocampus proBDNF as well as mature BDNF have been reported to be involved in hippocampal synaptic plasticity [299, 300]. Furthermore, secretion of proBDNF by activated microglia [250, 251] and expression of the proBDNF receptor

Mol Neurobiol

autophosphorylation of tyrosine residues within the intracellular part of the receptor creating docking sites for various adaptor proteins. Phosphorylation of tyrosine residue Y816 enables docking of PLC-γ and phosphorylation of tyrosine residue Y515 enables docking of Src homology 2 domain containing transforming protein (Shc) and FGF receptor substrate 2 (FRS-2), all of which activate various signal cascades ultimately leading to decreased KCC2 levels [308] through yet unknown cellular sorting mechanisms (Fig. 5). Consequently, chloride ions accumulate in the neurons and neuronal hyperpolarization via activated GABA(A) receptors can no longer be sustained [302, 303, 309, 310]. Therapeutic Approaches in the Spinal Cord to Inhibit Neuropathic Pain Neuronal hypersensitivity induced by nerve injury is a complex process initiated and maintained by interplay of spinal cord substances released by neurons and activated glial cells. Various therapeutic approaches are being explored in order to reverse injury-induced hypersensitivity. Glial Inhibition Activation of microglial P2X4 receptors appears to play a critical role in tactile allodynia development as inhibition of P2X4 receptors has been reported to impair microglial BDNF release and to reverse tactile allodynia [251, 271]. Furthermore, it has been suggested that BDNF affects astrocyte activation [311]. This suggests a treatment strategy based on inhibition of BDNF release from microglia upon nerve injury, e.g., via a P2X4 receptor antagonist. As gliosis is only triggered by abnormal events such as nerve injury, modulation of glial responses to restore normal neuronal patterns might be a possible treatment method without interfering with normal neuronal function. TrkB Antagonists Because activated microglia respond by releasing BDNF, thereby activating neuronal TrkB with consequent KCC2 downregulation, TrkB can be considered a potential drug target. In accordance, systemic administration of the TrkB antagonist cyclotraxin-B has been proven to prevent and reverse cold allodynia in animal models [312]. However, the concurrent inhibition of systemic normal TrkB function may prove to result in numerous side effects.

Conclusion Neurotrophin signaling is essential for key phases of PNS development, including cellular survival and differentiation,

neuronal sprouting and for the myelination process. Following injury to peripheral nerves, neurotrophin signaling pathways are reactivated in neurons and glia to facilitate nerve regeneration and functional recovery. Unfortunately, nerve recovery is often far from complete, and unwanted side effects of neurotrophin signaling in the spinal cord further initiate processes that lead to chronic neuropathic pain symptoms. Thus, there is an immense need to increase our understanding of fundamental biological aspects of neurotrophins and their receptors in neuronal and glial biology as well as their involvement in cellular communication. Hopefully, future research will facilitate the development of better treatments to increase functional nerve recovery after injury and to prevent or alleviate neuropathic pain. Acknowledgments The authors thank the Lundbeck Foundation, Simon Fougner Hartmanns Familiefond, Familien Hede-Nielsens Fond, Aase og Ejnar Danielsens Fond and PAINCAGE (European Framework Programme 7).

References 1. Battiston B, Papalia I, Tos P, Geuna S (2009) Chapter 1: Peripheral nerve repair and regeneration research: a historical note. Int Rev Neurobiol 87:1–7. doi:10.1016/S0074-7742(09)87001-3 2. NIH Publication No. 04-4853 (Peripheral Neuropathy Fact Sheet) 3. Chao MV (2003) Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 4:299–309. doi:10.1038/nrn1078 4. Kashiba H, Noguchi K, Ueda Y, Senba E (1995) Coexpression of trk family members and low-affinity neurotrophin receptors in rat dorsal root ganglion neurons. Brain Res Mol Brain Res 30:158–164 5. Wright DE, Snider WD (1995) Neurotrophin receptor mRNA expression defines distinct populations of neurons in rat dorsal root ganglia. J Comp Neurol 351:329–338 6. Bibel M, Hoppe E, Barde YA (1999) Biochemical and functional interactions between the neurotrophin receptors trk and p75NTR. EMBO J 18:616–622. doi:10.1093/emboj/18.3.616 7. Benedetti M, Levi A, Chao MV (1993) Differential expression of nerve growth factor receptors leads to altered binding affinity and neurotrophin responsiveness. Proc Natl Acad Sci U S A 90:7859– 7863 8. Hempstead BL, Martin-Zanca D, Kaplan DR et al (1991) Highaffinity NGF binding requires coexpression of the trk protooncogene and the low-affinity NGF receptor. Nature 350:678– 683. doi:10.1038/350678a0 9. Reichardt LF (2006) Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci 361:1545–1564. doi:10.1098/ rstb.2006.1894 10. Nykjaer A, Lee R, Teng KK et al (2004) Sortilin is essential for proNGF-induced neuronal cell death. Nature 427:843–848. doi:10. 1038/nature02319 11. Jansen P, Giehl K, Nyengaard JR et al (2007) Roles for the proneurotrophin receptor sortilin in neuronal development, aging and brain injury. Nat Neurosci 10:1449–1457. doi:10.1038/nn2000 12. Lee R, Kermani P, Teng KK, Hempstead BL (2001) Regulation of cell survival by secreted proneurotrophins. Science (New York, NY) 294:1945–1948. doi:10.1126/science.1065057

Mol Neurobiol 13. Masoudi R, Ioannou MS, Coughlin MD et al (2009) Biological activity of nerve growth factor precursor is dependent upon relative levels of its receptors. J Biol Chem 284:18424–18433. doi:10.1074/ jbc.M109.007104 14. Teng HK, Teng KK, Lee R et al (2005) ProBDNF induces neuronal apoptosis via activation of a receptor complex of p75NTR and sortilin. J Neurosci 25:5455–5463. doi:10.1523/JNEUROSCI. 5123-04.2005 15. Nikoletopoulou V, Lickert H, Frade JM et al (2010) Neurotrophin receptors TrkA and TrkC cause neuronal death whereas TrkB does not. Nature 467:59–63. doi:10.1038/nature09336 16. Martini FH (2005) Anatomy and physiology, 1st ed. Pearson Education, Inc 17. Richner M, Bjerrum OJ, Nykjaer A, Vaegter CB (2011) The Spared Nerve Injury (SNI) model of induced mechanical allodynia in mice. J Vis Exp. doi:10.3791/3092 18. Rigaud M, Gemes G, Barabas M-E et al (2008) Species and strain differences in rodent sciatic nerve anatomy: implications for studies of neuropathic pain. Pain 136:188–201. doi:10.1016/j.pain.2008.01.016 19. Amir R, Devor M (2003) Electrical excitability of the soma of sensory neurons is required for spike invasion of the soma, but not for through-conduction. Biophys J 84:2181–2191 20. Huang L-YM, Gu Y, Chen Y (2013) Communication between neuronal somata and satellite glial cells in sensory ganglia. Glia 1–11. doi: 10.1002/glia.22541 21. Pannese E (2010) The structure of the perineuronal sheath of satellite glial cells (SGCs) in sensory ganglia. Neuron Glia Biol 6: 3–10. doi:10.1017/S1740925X10000037 22. Pannese E, Ledda M, Arcidiacono G, Rigamonti L (1991) Clusters of nerve cell bodies enclosed within a common connective tissue envelope in the spinal ganglia of the lizard and rat. Cell Tissue Res 264:209–214 23. Pannese E (1981) The satellite cells of the sensory ganglia. Adv Anat Embryol Cell Biol 65:1–111 24. Hanani M (2005) Satellite glial cells in sensory ganglia: from form to function. Brain Res Brain Res Rev 48:457–476. doi:10.1016/j. brainresrev.2004.09.001 25. Jessen KR, Mirsky R (2005) The origin and development of glial cells in peripheral nerves. Nat Rev Neurosci 6:671–682. doi:10. 1038/nrn1746 26. Bear MF, Connors BW, Paradiso MA (2007) Neuroscience: exploring the brain, 3rd edn. Lippincott Williams & Wilkins, Baltimore, MD 27. Manzano GM, Giuliano LMP, Nóbrega JAM (2008) A brief historical note on the classification of nerve fibers. Arq Neuropsiquiatr 66: 117–119 28. Lewin GR, Moshourab R (2004) Mechanosensation and pain. J Neurobiol 61:30–44. doi:10.1002/neu.20078 29. Lallemend F, Ernfors P (2012) Molecular interactions underlying the specification of sensory neurons. Trends Neurosci 35:373–381. doi:10.1016/j.tins.2012.03.006 30. Honma Y, Kawano M, Kohsaka S, Ogawa M (2010) Axonal projections of mechanoreceptive dorsal root ganglion neurons depend on Ret. Development (Cambridge, England) 137:2319–2328. doi:10.1242/dev.046995 31. Flemming W (1882) Flemming: Vom Bau der Spinalganglienzellen. Festschrift für Henle 32. Daae H (1887) Zur Kenntniss der Spinalganglienzellen beim Säugethier. Arch Mikrosk Anat 31:223–235. doi:10.1007/ BF02955708 33. Müller E (1891) Untersuchungen über den Bau der Spinal-ganglien. Nordiskt Medicinskt Arkiv 23:1–55. doi:10.1111/j.0954-6820. 1891.tb00764.x 34. Lawson SN (1979) The postnatal development of large light and small dark neurons in mouse dorsal root ganglia: a statistical analysis of cell numbers and size. J Neurocytol 8:275–294

35. Hossack J, Wyburn GM (1954) XVI.—Electron Microscopic Studies of Spinal Ganglion Cells. Proc R Soc Edinb B Biol 65: 239–250. doi:10.1017/S0080455X00012133 36. Andres KH (1961) Untersuchungen über den Feinbau von Spinalganglien. Cell Tissue Res 55:1–48. doi:10.1007/ BF00319327 37. Lawson SN, Harper AA, Harper EI et al (1984) A monoclonal antibody against neurofilament protein specifically labels a subpopulation of rat sensory neurones. J Comp Neurol 228:263–272. doi: 10.1002/cne.902280211 3 8 . Av e r i l l S , M c M a h o n S B , C l a r y D O e t a l ( 1 9 9 5 ) Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur J Neurosci 7: 1484–1494. doi:10.1111/j.1460-9568.1995.tb01143.x 39. Priestley JV, Michael GJ, Averill S et al (2002) Regulation of nociceptive neurons by nerve growth factor and glial cell line derived neurotrophic factor. Can J Physiol Pharmacol 80:495– 505. doi:10.1139/y02-034 40. Scott SA (1992) Sensory neurons: diversity, development, and plasticity. Oxford University Press 41. Price J (1985) An immunohistochemical and quantitative examination of dorsal root ganglion neuronal subpopulations. J Neurosci 5: 2051–2059 42. Harper AA, Lawson SN (1985) Conduction velocity is related to morphological cell type in rat dorsal root ganglion neurones. J Physiol Lond 359:31–46 43. Karchewski LA, Kim FA, Johnston J et al (1999) Anatomical evidence supporting the potential for modulation by multiple neurotrophins in the majority of adult lumbar sensory neurons. J Comp Neurol 413:327–341 44. Verge VM, Merlio JP, Grondin J et al (1992) Colocalization of NGF binding sites, trk mRNA, and low-affinity NGF receptor mRNA in primary sensory neurons: responses to injury and infusion of NGF. J Neurosci 12:4011–4022 45. Verge VMK, Richardson PM, Benoit R, Riopelle RJ (1989) Histochemical characterization of sensory neurons with highaffinity receptors for nerve growth factor. J Neurocytol 18:583– 591. doi:10.1007/BF01187079 46. Mu X, Silos-Santiago I, Carroll SL, Snider WD (1993) Neurotrophin receptor genes are expressed in distinct patterns in developing dorsal root ganglia. J Neurosci 13:4029–4041 47. Bennett DLD, Averill SS, Clary DOD et al (1996) Postnatal changes in the expression of the trkA high-affinity NGF receptor in primary sensory neurons. Eur J Neurosci 8:2204–2208. doi:10.1111/j.14609568.1996.tb00742.x 48. McMahon SB, Armanini MP, Ling LH, Phillips HS (1994) Expression and coexpression of Trk receptors in subpopulations of adult primary sensory neurons projecting to identified peripheral targets. Neuron 12:1161–1171 49. Molliver DC, Radeke MJ, Feinstein SC, Snider WD (1995) Presence or absence of TrKA protein distinguishes subsets of small sensory neurons with unique cytochemical characteristics and dorsal horn projections. J Comp Neurol 361:404–416. doi:10.1002/ cne.903610305 50. Smeyne RJ, Klein R, Schnapp A et al (1994) Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene. Nature 368:246–249. doi:10.1038/ 368246a0 51. Crowley C, Spencer SD, Nishimura MC et al (1994) Mice lacking nerve growth factor display perinatal loss of sensory and sympathetic neurons yet develop basal forebrain cholinergic neurons. Cell 76:1001–1011 52. Stucky CL, Koltzenburg M, Schneider M et al (1999) Overexpression of nerve growth factor in skin selectively affects the survival and functional properties of nociceptors. J Neurosci 19: 8509–8516

Mol Neurobiol 53. Wetmore C, Olson L (1995) Neuronal and nonneuronal expression of neurotrophins and their receptors in sensory and sympathetic ganglia suggest new intercellular trophic interactions. J Comp Neurol 353:143–159. doi:10.1002/cne.903530113 54. Michael GJG, Averill SS, Nitkunan AA et al (1997) Nerve growth factor treatment increases brain-derived neurotrophic factor selectively in TrkA-expressing dorsal root ganglion cells and in their central terminations within the spinal cord. J Neurosci 17:8476– 8490 55. Snider WD (1994) Functions of the neurotrophins during nervous system development: what the knockouts are teaching us. Cell 77: 627–638 56. Bennett DL, Michael GJ, Ramachandran N et al (1998) A distinct subgroup of small DRG cells express GDNF receptor components and GDNF is protective for these neurons after nerve injury. J Neurosci 18:3059–3072 57. Snider WD, McMahon SB (1998) Tackling pain at the source: new ideas about nociceptors. Neuron 20:629–632 58. Light AR, Trevino DL, Perl ER (1979) Morphological features of functionally defined neurons in the marginal zone and substantia gelatinosa of the spinal dorsal horn. J Comp Neurol 186:151–171. doi:10.1002/cne.901860204 59. Malmberg AB (1997) Preserved acute pain and reduced neuropathic pain in mice lacking PKC. Science (New York, NY) 278:279–283. doi:10.1126/science.278.5336.279 60. Pannese E, Procacci P (2002) Ultrastructural localization of NGF receptors in satellite cells of the rat spinal ganglia. J Neurocytol 31: 755–763. doi:10.1023/A:1025708132119 61. Kuo L-T, Groves MJ, Scaravilli F et al (2007) Neurotrophin-3 administration alters neurotrophin, neurotrophin receptor and nestin mRNA expression in rat dorsal root ganglia following axotomy. Neuroscience 147:491–507. doi:10.1016/j.neuroscience.2007.04.023 62. Zhou X-F, Chie ET, Deng YS et al (1999) Injured primary sensory neurons switch phenotype for brain-derived neurotrophic factor in the rat. Neuroscience 92:841–853 63. Zhou X-F, Deng YS, Chie E et al (1999) Satellite-cell-derived nerve growth factor and neurotrophin-3 are involved in noradrenergic sprouting in the dorsal root ganglia following peripheral nerve injury in the rat. Eur J Neurosci 11:1711–1722 64. van Velzen M, Laman JD, Kleinjan A et al (2009) Neuroninteracting satellite glial cells in human trigeminal ganglia have an APC phenotype. J Immunol 183:2456–2461. doi:10.4049/ jimmunol.0900890 65. Geuna S, Raimondo S, Ronchi G et al (2009) Chapter 3: Histology of the peripheral nerve and changes occurring during nerve regeneration. Int Rev Neurobiol 87:27–46. doi:10.1016/S0074-7742(09) 87003-7 66. Geuna S, Fornaro M, Raimondo S, Giacobini-Robecchi MG (2010) Plasticity and regeneration in the peripheral nervous system. Ital J Anat Embryol 115:91–94 67. Huang T-Y, Belzer V, Hanani M (2010) Gap junctions in dorsal root ganglia: possible contribution to visceral pain. Eur J Pain 14(49): e1–11. doi:10.1016/j.ejpain.2009.02.005 68. Ramer MS, Bradbury EJ (2001) Nerve growth factor induces P2X3 expression in sensory neurons. J Neurochem 77:864–875 69. Hanani M, Huang TY, Cherkas PS et al (2002) Glial cell plasticity in sensory ganglia induced by nerve damage. Neuroscience 114:279– 283 70. Pannese E, Ledda M, Cherkas PS et al (2003) Satellite cell reactions to axon injury of sensory ganglion neurons: increase in number of gap junctions and formation of bridges connecting previously separate perineuronal sheaths. Anat Embryol 206:337–347. doi:10. 1007/s00429-002-0301-6 71. Bergman E, Fundin BT, Ulfhake B (1999) Effects of aging and axotomy on the expression of neurotrophin receptors in primary sensory neurons. J Comp Neurol 410:368–386

72. Obata K, Katsura H, Sakurai J et al (2006) Suppression of the p75 neurotrophin receptor in uninjured sensory neurons reduces neuropathic pain after nerve injury. J Neurosci 26:11974–11986. doi:10. 1523/JNEUROSCI.3188-06.2006 73. Apfel SC, Wright DE, Wiideman AM et al (1996) Nerve growth factor regulates the expression of brain-derived neurotrophic factor mRNA in the peripheral nervous system. Mol Cell Neurosci 7:134– 142. doi:10.1006/mcne.1996.0010 74. Cho HJH, Kim JKJ, Park HCH et al (1998) Changes in brainderived neurotrophic factor immunoreactivity in rat dorsal root ganglia, spinal cord, and gracile nuclei following cut or crush injuries. Exp Neurol 154:7–7. doi:10.1006/exnr.1998.6936 75. Fukuoka T, Kondo E, Dai Y et al (2001) Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model. J Neurosci 21:4891–4900 76. Mannion RJ, Costigan M, Decosterd I et al (1999) Neurotrophins: peripherally and centrally acting modulators of tactile stimulusinduced inflammatory pain hypersensitivity. Proc Natl Acad Sci U S A 96:9385–9390. doi:10.1073/pnas.96.16.9385 77. Bennett DL (2001) Neurotrophic factors: important regulators of nociceptive function. Neuroscientist 7:13–17 78. Pezet S, Malcangio M, Lever IJ et al (2002) Noxious stimulation induces Trk receptor and downstream ERK phosphorylation in spinal dorsal horn. Mol Cell Neurosci 21:684–695 79. McMahon SBS, Jones NGN (2004) Plasticity of pain signaling: role of neurotrophic factors exemplified by acid-induced pain. J Neurobiol 61:72–87. doi:10.1002/neu.20093 80. Ha SOS, Kim JKJ, Hong HSH et al (2000) Expression of brainderived neurotrophic factor in rat dorsal root ganglia, spinal cord and gracile nuclei in experimental models of neuropathic pain. Neuroscience 107:301–309 81. Ramer MS, Thompson SW, McMahon SB (1999) Causes and consequences of sympathetic basket formation in dorsal root ganglia. Pain Suppl 6:S111–20 82. Pezet S, McMahon SB (2006) Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci 29:507–538. doi:10.1146/ annurev.neuro.29.051605.112929 83. Cajal SRY (1928) Degeneration and regeneration of the nervous system. Oxford University Press, Oxford 84. Munson JB, Shelton DL, McMahon SB (1997) Adult mammalian sensory and motor neurons: roles of endogenous neurotrophins and rescue by exogenous neurotrophins after axotomy. J Neurosci 17: 470–476 85. Kurata S, Goto T, Gunjigake KK et al (2013) Nerve growth factor involves mutual interaction between neurons and satellite glial cells in the rat trigeminal ganglion. Acta Histochem Cytochem 46:65–73. doi:10.1267/ahc.13003 86. Castillo C, Norcini M, Martin Hernandez LA et al (2013) Satellite glia cells in dorsal root ganglia express functional NMDA receptors. Neuroscience 240C:135–146. doi:10.1016/j.neuroscience.2013.02. 031 87. Dublin P, Hanani M (2007) Satellite glial cells in sensory ganglia: their possible contribution to inflammatory pain. Brain Behav Immun 21:592–598. doi:10.1016/j.bbi.2006.11.011 88. Ohara PT, Vit J-P, Bhargava A et al (2009) Gliopathic pain: when satellite glial cells go bad. Neuroscientist 15:450–463. doi:10.1177/ 1073858409336094 89. Jasmin L, Vit J-P, Bhargava A, Ohara PT (2010) Can satellite glial cells be therapeutic targets for pain control? Neuron Glia Biol 6:63– 71. doi:10.1017/S1740925X10000098 90. Qiao L-YL, Vizzard MAM (2005) Spinal cord injury-induced expression of TrkA, TrkB, phosphorylated CREB, and c-Jun in rat lumbosacral dorsal root ganglia. J Comp Neurol 482:142–154. doi: 10.1002/cne.20394 91. Miller FD, Speelman A, Mathew TC et al (1994) Nerve growth factor derived from terminals selectively increases the ratio of p75 to

Mol Neurobiol

92.

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

105.

106.

107.

trkA NGF receptors on mature sympathetic neurons. Dev Biol 161: 206–217. doi:10.1006/dbio.1994.1021 Belliveau DJ (1997) NGF and neurotrophin-3 both activate TrkA on sympathetic neurons but differentially regulate survival and neuritogenesis. J Cell Biol 136:375–388. doi:10.1083/jcb.136.2. 375 Zhou X-F, Rush RA, McLachlan EM (1996) Differential expression of the p75 nerve growth factor receptor in glia and neurons of the rat dorsal root ganglia after peripheral nerve transection. J Neurosci 16: 2901–2911 Ramer M, Bisby M (1997) Reduced sympathetic sprouting occurs in dorsal root ganglia after axotomy in mice lacking low-affinity neurotrophin receptor. Neurosci Lett 228:9–12 Otto DD, Unsicker KK, Grothe CC (1987) Pharmacological effects of nerve growth factor and fibroblast growth factor applied to the transectioned sciatic nerve on neuron death in adult rat dorsal root ganglia. Neurosci Lett 83:156–160 Cheema SS, Barrett GL, Bartlett PF (1996) Reducing p75 nerve growth factor receptor levels using antisense oligonucleotides prevents the loss of axotomized sensory neurons in the dorsal root ganglia of newborn rats. J Neurosci Res 46:239–245. doi:10.1002/ (SICI)1097-4547(19961015)46:23.0.CO;2-Y Vestergaard S, Tandrup T, Jakobsen J (1997) Effect of permanent axotomy on number and volume of dorsal root ganglion cell bodies. J Comp Neurol 388:307–312 Degn J, Tandrup T, Jakobsen J (1999) Effect of nerve crush on perikaryal number and volume of neurons in adult rat dorsal root ganglion. J Comp Neurol 412:186–192. doi:10.1002/(SICI)10969861(19990913)412:13.0.CO;2-H Shi TJ, Tandrup T, Bergman E et al (2001) Effect of peripheral nerve injury on dorsal root ganglion neurons in the C57 BL/6 J mouse: marked changes both in cell numbers and neuropeptide expression. Neuroscience 105:249–263 Gjerstad MD, Tandrup T, Koltzenburg M, Jakobsen J (2002) Predominant neuronal B-cell loss in L5 DRG of p75 receptordeficient mice. J Anat 200:81–87 Sørensen B, Tandrup T, Koltzenburg M, Jakobsen J (2003) No further loss of dorsal root ganglion cells after axotomy in p75 neurotrophin receptor knockout mice. J Comp Neurol 459:242– 250. doi:10.1002/cne.10625 Zhou X-F, Li W-P, Zhou FH-H et al (2005) Differential effects of endogenous brain-derived neurotrophic factor on the survival of axotomized sensory neurons in dorsal root ganglia: a possible role for the p75 neurotrophin receptor. Neuroscience 132:591–603. doi: 10.1016/j.neuroscience.2004.12.034 Jiang Y, Zhang JS, Jakobsen J (2005) Differential effect of p75 neurotrophin receptor on expression of pro-apoptotic proteins c-jun, p38 and caspase-3 in dorsal root ganglion cells after axotomy in experimental diabetes. Neuroscience 132:1083–1092. doi:10.1016/ j.neuroscience.2005.01.049 Jiang Y, Jakobsen J (2010) Different apoptotic reactions of dorsal root ganglion A- and B-cells after sciatic nerve axotomy: effect of p75 neurotrophin receptor. Chin Med J 123:2695– 2700 Arnett MG, Ryals JM, Wright DE (2007) Pro-NGF, sortilin, and p75NTR: potential mediators of injury-induced apoptosis in the mouse dorsal root ganglion. Brain Res Rev 1183:32–42. doi:10. 1016/j.brainres.2007.09.051 Vaegter CB, Jansen P, Fjorback AW et al (2011) Sortilin associates with Trk receptors to enhance anterograde transport and neurotrophin signaling. Nat Neurosci 14:54–61. doi:10.1038/nn.2689 Averill SS, Michael GJG, Shortland PJP et al (2004) NGF and GDNF ameliorate the increase in ATF3 expression which occurs in dorsal root ganglion cells in response to peripheral nerve injury. Eur J Neurosci 19:1437–1445. doi:10.1111/j.1460-9568.2004. 03241.x

108. Zwick M, Davis BM, Woodbury CJ et al (2002) Glial cell linederived neurotrophic factor is a survival factor for isolectin B4positive, but not vanilloid receptor 1-positive, neurons in the mouse. J Neurosci 22:4057–4065 109. Obata KK, Yamanaka HH, Kobayashi KK et al (2004) Role of mitogen-activated protein kinase activation in injured and intact primary afferent neurons for mechanical and heat hypersensitivity after spinal nerve ligation. J Neurosci 24:10211–10222. doi:10. 1523/JNEUROSCI.3388-04.2004 110. Obata K, Yamanaka H, Dai Y et al (2004) Differential activation of MAPK in injured and uninjured DRG neurons following chronic constriction injury of the sciatic nerve in rats. Eur J Neurosci 20: 2881–2895. doi:10.1111/j.1460-9568.2004.03754.x 111. Bergmann I, Reiter R, Toyka KV, Koltzenburg M (1998) Nerve growth factor evokes hyperalgesia in mice lacking the low-affinity neurotrophin receptor p75. Neurosci Lett 255:87–90 112. Chuang HH, Prescott ED, Kong H et al (2001) Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4, 5)P2-mediated inhibition. Nature 411:957–962. doi:10.1038/ 35082088 113. Bonnington JKJ, McNaughton PAP (2003) Signalling pathways involved in the sensitisation of mouse nociceptive neurones by nerve growth factor. J Physiol 551:433–446. doi:10.1113/jphysiol. 2003.039990 114. Kobayashi K, Fukuoka T, Obata K et al (2005) Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with adelta/c-fibers and colocalization with trk receptors. J Comp Neurol 493:596–606. doi:10.1002/cne.20794 115. Ji R-R, Samad TA, Jin S-X et al (2002) p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 36:57–68 116. Bron R, Klesse LJ, Shah K et al (2003) Activation of Ras is necessary and sufficient for upregulation of vanilloid receptor type 1 in sensory neurons by neurotrophic factors. Mol Cell Neurosci 22:118–132 117. Kerr BJ, Souslova V, McMahon SB, Wood JN (2001) A role for the TTX-resistant sodium channel Nav 1.8 in NGF-induced hyperalgesia, but not neuropathic pain. Neuroreport 12:3077–3080 118. Mamet J, Lazdunski M, Voilley N (2003) How nerve growth factor drives physiological and inflammatory expressions of acid-sensing ion channel 3 in sensory neurons. J Biol Chem 278:48907–48913. doi:10.1074/jbc.M309468200 119. Mantyh PW, Koltzenburg M, Mendell LM et al (2011) Antagonism of nerve growth factor-TrkA signaling and the relief of pain. Anesthesiology 115:189–204. doi:10.1097/ALN.0b013e31821b1ac5 120. Thomas PK, Berthold CH, Ochoa J (1993) Microscopic anatomy of the peripheral nervous system. In: Peripheral neuropathy, 3rd ed. WB Saunders, Philadelphia, PA, pp 28–92 121. Key A, Retzius G (1876) Studien der Anatomie des Nervensystems und des Bindegewebes. Samson & Wallin 122. Akert K, Sandri C, Weibel ER et al (1976) The fine structure of the perineural endothelium. Cell Tissue Res 165:281–295 123. Gaudet AD, Popovich PG, Ramer MS (2011) Wallerian degeneration: gaining perspective on inflammatory events after peripheral nerve injury. J Neuroinflamm 8:110. doi:10.1523/JNEUROSCI. 2269-10.2010 124. Lubińska L (1982) Patterns of Wallerian degeneration of myelinated fibres in short and long peripheral stumps and in isolated segments of rat phrenic nerve. Interpretation of the role of axoplasmic flow of the trophic factor. Brain Res Rev 233:227–240 125. Schlaepfer WW (1977) Structural alterations of peripheral nerve induced by the calcium ionophore A23187. Brain Res Rev 136:1–9 126. Vial JD (1958) The early changes in the axoplasm during Wallerian degeneration. J Biophys Biochem Cytol 4:551–555 127. Ghabriel MN, Allt G (1979) The role of Schmidt–Lanterman incisures in Wallerian degeneration: II. An electron microscopic study. Acta Neuropathol 48:95–103

Mol Neurobiol 128. Yao D, Li M, Shen D et al (2013) Expression changes and bioinformatic analysis of Wallerian degeneration after sciatic nerve injury in rat. Neurosci Bull 29:321–332. doi:10.1007/s12264-013-1340-0 129. Funakoshi H, Risling M, Carlstedt T, et al. (1998) Targeted expression of a multifunctional chimeric neurotrophin in the lesioned sciatic nerve accelerates regeneration of sensory and motor axons. 95:5269–5274 130. Funakoshi H, Frisén J, Barbany G et al (1993) Differential expression of mRNAs for neurotrophins and their receptors after axotomy of the sciatic nerve. J Cell Biol 123:455–465 131. Heumann R, Lindholm D, Bandtlow C et al (1987) Differential regulation of mRNA encoding nerve growth factor and its receptor in rat sciatic nerve during development, degeneration, and regeneration: role of macrophages. Proc Natl Acad Sci U S A 84:8735–8739 132. Thoenen H, Bandtlow C, Heumann R et al (1988) Nerve growth factor: cellular localization and regulation of synthesis. Cell Mol Neurobiol 8:35–40 133. Warner LE, Mancias P, Butler IJ et al (1998) Mutations in the early growth response 2 (EGR2) gene are associated with hereditary myelinopathies. Nat Genet 18:382–384. doi:10.1038/ng0498-382 134. Jessen KR, Mirsky R (2008) Negative regulation of myelination: relevance for development, injury, and demyelinating disease. Glia 56:1552–1565. doi:10.1002/glia.20761 135. Fontana X, Hristova M, Da Costa C et al (2012) c-Jun in Schwann cells promotes axonal regeneration and motoneuron survival via paracrine signaling. J Cell Biol 198:127–141. doi:10.1083/jcb. 201205025 136. Kim CF, Moalem-Taylor G (2011) Detailed characterization of neuro-immune responses following neuropathic injury in mice. Brain Res Rev 1405:95–108. doi:10.1016/j.brainres.2011.06.022 137. Shubayev VI, Angert M, Dolkas J et al (2006) TNFalpha-induced MMP-9 promotes macrophage recruitment into injured peripheral nerve. Mol Cell Neurosci 31:407–415. doi:10.1016/j.mcn.2005.10. 011 138. Dubový P (2011) Wallerian degeneration and peripheral nerve conditions for both axonal regeneration and neuropathic pain induction. Ann Anat 193:267–275. doi:10.1016/j.aanat.2011.02.011 139. Chen Z-L, Yu W-M, Strickland S (2007) Peripheral regeneration. Annu Rev Neurosci 30:209–33. doi:10.1146/annurev.neuro.30. 051606.094337 140. Fu SY, Gordon T (1997) The cellular and molecular basis of peripheral nerve regeneration. Mol Neurobiol 14:67–116. doi:10. 1007/BF02740621 141. Sunderland S (1952) Factors influencing the course of regeneration and the quality of the recovery after nerve suture. Brain 75:19–54 142. Ernfors P, Lee K-F, Jaenisch R (1994) Mice lacking brain-derived neurotrophic factor develop with sensory deficits. Nature 368:147– 150. doi:10.1038/368147a0 143. Fariñas I, Jones KR, Backus C et al (1994) Severe sensory and sympathetic deficits in mice lacking neurotrophin-3. Nature 369: 658–661. doi:10.1038/369658a0 144. Yan Q, Elliott J, Snider WD (1992) Brain-derived neurotrophic factor rescues spinal motor neurons from axotomy-induced cell death. Nature 360:753–5. doi:10.1038/360753a0 145. Sendtner M, Holtmann B, Kolbeck R et al (1992) Brain-derived neurotrophic factor prevents the death of motoneurons in newborn rats after nerve section. Nature 360:757–759. doi:10.1038/ 360757a0 146. Koliatsos VE, Clatterbuck RE, Winslow JW et al (1993) Evidence that brain-derived neurotrophic factor is a trophic factor for motor neurons in vivo. Neuron 10:359–367 147. Funakoshi H, Belluardo N, Arenas E, et al. (1995) Muscle-derived neurotrophin-4 as an activity-dependent trophic signal for adult motor neurons. Science (New York, NY) 268:1495–1499 148. Meyer M, Matsuoka I, Wetmore C et al (1992) Enhanced synthesis of brain-derived neurotrophic factor in the lesioned peripheral

nerve: different mechanisms are responsible for the regulation of BDNF and NGF mRNA. J Cell Biol 119:45–54 149. Zhang JYJ, Luo XGX, Xian CJC et al (2000) Endogenous BDNF is required for myelination and regeneration of injured sciatic nerve in rodents. Eur J Neurosci 12:4171–4180. doi:10.1111/j.1460-9568. 2000.01312.x 150. Acheson A, Barker PA, Alderson RF et al (1991) Detection of brain-derived neurotrophic factor-like activity in fibroblasts and Schwann cells: inhibition by antibodies to NGF. Neuron 7:265–275 151. Zhou X-F, Rush RA (1996) Endogenous brain-derived neurotrophic factor is anterogradely transported in primary sensory neurons. Neuroscience 74:945–953 152. Tonra JR, Curtis R, Wong V et al (1998) Axotomy upregulates the anterograde transport and expression of brain-derived neurotrophic factor by sensory neurons. J Neurosci 18:4374–4383 153. Korte M, Carroll P, Wolf E et al (1995) Hippocampal long-term potentiation is impaired in mice lacking brain-derived neurotrophic factor. Proc Natl Acad Sci U S A 92:8856–8860 154. English AW, Meador W, Carrasco DI (2005) Neurotrophin-4/5 is required for the early growth of regenerating axons in peripheral nerves. Eur J Neurosci 21:2624–2634. doi:10.1111/j.1460-9568. 2005.04124.x 155. Boyd JG, Gordon T (2002) A dose-dependent facilitation and inhibition of peripheral nerve regeneration by brain-derived neurotrophic factor. Eur J Neurosci 15:613–626 156. Fu SY, Gordon T (1995) Contributing factors to poor functional recovery after delayed nerve repair: prolonged denervation. J Neurosci 15:3886–3895 157. Wood SJ, Pritchard J, Sofroniew MV (1989) Re-expression of nerve growth factor receptor after axonal injury recapitulates a developmental event in motor neurons: differential regulation when regeneration is allowed or prevented. Eur J Neurosci 2:650–657. doi:10. 1111/j.1460-9568.1990.tb00454.x 158. Ernfors P, Henschen A, Olson L, Persson H (1989) Expression of nerve growth factor receptor mRNA is developmentally regulated and increased after axotomy in rat spinal cord motoneurons. Neuron 2:1605–1613 159. Bussmann KAV, Sofroniew MV (1999) Re-expression of p75NTR by adult motor neurons after axotomy is triggered by retrograde transport of a positive signal from axons regrowing through damaged or denervated peripheral nerve tissue. Neuroscience 91:273– 281. doi:10.1016/S0306-4522(98)00562-4 160. Taniuchi M, Clark HB, Schweitzer JB, Johnson EM Jr (1988) Expression of nerve growth factor receptors by Schwann cells of axotomized peripheral nerves: ultrastructural location, suppression by axonal contact, and binding properties. J Neurosci 8: 664–681 161. Taniuchi M, Clark HB, Johnson EM Jr (1986) Induction of nerve growth factor receptor in Schwann cells after axotomy. Proc Natl Acad Sci U S A 83:4094–4098 162. Tomita K, Kubo T, Matsuda K et al (2007) The neurotrophin receptor p75NTR in Schwann cells is implicated in remyelination and motor recovery after peripheral nerve injury. Glia 55:1199– 1208. doi:10.1002/glia.20533 163. Wilhelm JC, Xu M, Cucoranu D et al (2012) Cooperative roles of BDNF expression in neurons and Schwann cells are modulated by exercise to facilitate nerve regeneration. J Neurosci 32:5002–5009. doi:10.1523/JNEUROSCI.1411-11.2012 164. Geremia NM, Pettersson LME, Hasmatali JC et al (2010) Endogenous BDNF regulates induction of intrinsic neuronal growth programs in injured sensory neurons. Exp Neurol 223: 128–142. doi:10.1016/j.expneurol.2009.07.022 165. Sun Y, Lim Y, Li F et al (2012) ProBDNF collapses neurite outgrowth of primary neurons by activating RhoA. PLoS ONE 7: e35883. doi:10.1371/journal.pone.0035883

Mol Neurobiol 166. Heumann R, Korsching S, Bandtlow C, Thoenen H (1987) Changes of nerve growth factor synthesis in nonneuronal cells in response to sciatic nerve transection. J Cell Biol 104:1623–1631 167. Lindholm D, Heumann R, Meyer M, Thoenen H (1987) Interleukin1 regulates synthesis of nerve growth factor in non-neuronal cells of rat sciatic nerve. Nature 330:658–659. doi:10.1038/330658a0 168. Johnson EM Jr, Taniuchi M, DiStefano PS (1988) Expression and possible function of nerve growth factor receptors on Schwann cells. Trends Neurosci 11:299–304 169. Diamond J, Foerster A, Holmes M, Coughlin M (1992) Sensory nerves in adult rats regenerate and restore sensory function to the skin independently of endogenous NGF. J Neurosci 12:1467–1476 170. Meier C, Parmantier E, Brennan A et al (1999) Developing Schwann cells acquire the ability to survive without axons by establishing an autocrine circuit involving insulin-like growth factor, neurotrophin-3, and platelet-derived growth factor-BB. J Neurosci 19:3847–3859 171. Sahenk Z, Oblinger J, Edwards C (2008) Neurotrophin-3 deficient Schwann cells impair nerve regeneration. Exp Neurol 212:552–6. doi:10.1016/j.expneurol.2008.04.015 172. Li H, Terenghi G, Hall SM (1997) Effects of delayed re-innervation on the expression of c-erbB receptors by chronically denervated rat Schwann cells in vivo. Glia 20:333–347 173. Sulaiman OA, Gordon T (2000) Effects of short- and long-term Schwann cell denervation on peripheral nerve regeneration, myelination, and size. Glia 32:234–246. doi:10.1002/10981136(200012)32:33.0.CO;2-3 174. Griesbeck O, Parsadanian AS, Sendtner M, Thoenen H (1995) Expression of neurotrophins in skeletal muscle: quantitative comparison and significance for motoneuron survival and maintenance of function. J Neurosci Res 42:21–33. doi:10.1002/jnr.490420104 175. Yin Q, kemp GJ, Yu L-G (2001) Neurotrophin-4 delivered by fibrin glue promotes peripheral nerve regeneration. Muscle Nerve 24: 345–351. doi:10.1002/1097-4598(200103)24:33.0.CO;2-P 176. Jessen KR, Mirsky R (2002) Signals that determine Schwann cell identity. J Anat 200:367–376 177. Höke A (2006) Schwann cells express motor and sensory phenotypes that regulate axon regeneration. J Neurosci 26:9646–9655. doi:10.1523/JNEUROSCI.1620-06.2006 178. Brushart TM, Aspalter M, Griffin JW et al (2013) Schwann cell phenotype is regulated by axon modality and central–peripheral location, and persists in vitro. Exp Neurol 247:272–281. doi:10. 1016/j.expneurol.2013.05.007 179. Meyer zu Hörste G, Prukop T, Nave K-A, Sereda MW (2006) Myelin disorders: causes and perspectives of Charcot–Marie– Tooth neuropathy. J Mol Neurosci 28:77–88 180. Tzakos AG, Troganis A, Theodorou V et al (2005) Structure and function of the myelin proteins: current status and perspectives in relation to multiple sclerosis. Curr Med Chem 12:1569–1587 181. Bunge MB, Williams AK, Wood PM (1982) Neuron–Schwann cell interaction in basal lamina formation. Dev Biol 92:449–460 182. Yamauchi J, Chan JR, Shooter EM (2003) Neurotrophin 3 activation of TrkC induces Schwann cell migration through the c-Jun Nterminal kinase pathway. Proc Natl Acad Sci U S A 100:14421– 14426. doi:10.1073/pnas.2336152100 183. Yamauchi J, Chan JR, Shooter EM (2004) Neurotrophins regulate Schwann cell migration by activating divergent signaling pathways dependent on Rho GTPases. Proc Natl Acad Sci U S A 101:8774– 8779. doi:10.1073/pnas.0402795101 184. Anton ES, Weskamp G, Reichardt LF, Matthew WD (1994) Nerve growth factor and its low-affinity receptor promote Schwann cell migration. Proc Natl Acad Sci U S A 91:2795–2799 185. Bentley CA, Lee KF (2000) p75 is important for axon growth and Schwann cell migration during development. J Neurosci 20:7706– 7715

186. Chan JR, Watkins TA, Cosgaya JM et al (2004) NGF controls axonal receptivity to myelination by Schwann cells or oligodendrocytes. Neuron 43:183–191 187. Chan JR, Cosgaya JM, Wu YJ, Shooter EM (2001) Neurotrophins are key mediators of the myelination program in the peripheral nervous system. Proc Natl Acad Sci U S A 98:14661–14668. doi: 10.1073/pnas.251543398 188. Ng BK, Chen L, Mandemakers W et al (2007) Anterograde transport and secretion of brain-derived neurotrophic factor along sensory axons promote Schwann cell myelination. J Neurosci 27:7597– 7603. doi:10.1523/JNEUROSCI.0563-07.2007 189. Cosgaya JM, Chan JR, Shooter EM (2002) The neurotrophin receptor p75NTR as a positive modulator of myelination. Science (New York, NY) 298:1245–1248. doi:10.1126/science.1076595 190. Song X-Y, Zhou FH-H, Zhong J-H et al (2006) Knockout of p75NTR impairs re-myelination of injured sciatic nerve in mice. J Neurochem 96:833–842. doi:10.1111/j.1471-4159.2005.03564.x 191. Kemp SWP, Webb AA, Dhaliwal S et al (2011) Dose and duration of nerve growth factor (NGF) administration determine the extent of behavioral recovery following peripheral nerve injury in the rat. Exp Neurol 229:460–470. doi:10.1016/j.expneurol.2011.03.017 192. Jubran M, Widenfalk J (2003) Repair of peripheral nerve transections with fibrin sealant containing neurotrophic factors. Exp Neurol 181:204–212 193. Tannemaat MR, Boer GJ, Verhaagen J, Malessy MJA (2007) Genetic modification of human surel nerve segments by a lentiviral vector encoding nerve growth factor. Neurosurgery 61:1286–1296. doi:10.1227/01.neu.0000306108.78044.a2 194. Tannemaat MR, Eggers R, Hendriks WT et al (2008) Differential effects of lentiviral vector-mediated overexpression of nerve growth factor and glial cell line-derived neurotrophic factor on regenerating sensory and motor axons in the transected peripheral nerve. Eur J Neurosci 28:1467–1479. doi:10.1111/j.1460-9568.2008.06452.x 195. Hu X, Cai J, Yang J, Smith GM (2010) Sensory axon targeting is increased by NGF gene therapy within the lesioned adult femoral nerve. Exp Neurol 223:153–165. doi:10.1016/j.expneurol.2009.08. 025 196. de Boer R, Knight AM, Borntraeger A et al (2011) Rat sciatic nerve repair with a poly-lactic-co-glycolic acid scaffold and nerve growth factor releasing microspheres. Microsurgery 31:293–302. doi:10. 1002/micr.20869 197. Chung T-W, Yang M-C, Tseng C-C et al (2011) Promoting regeneration of peripheral nerves in-vivo using new PCL-NGF/Tirofiban nerve conduits. Biomaterials 32:734–743. doi:10.1016/j. biomaterials.2010.09.023 198. Yan Q, Yin Y, Li B (2012) Use new PLGL-RGD-NGF nerve conduits for promoting peripheral nerve regeneration. Biomed Eng Online 11:36. doi:10.1186/1475-925X-11-36 199. Shakhbazau A, Kawasoe J, Hoyng SA et al (2012) Early regenerative effects of NGF-transduced Schwann cells in peripheral nerve repair. Mol Cell Neurosci 50:103–112. doi:10.1016/j.mcn. 2012.04.004 200. Sahenk Z, Nagaraja HN, McCracken BS et al (2005) NT-3 promotes nerve regeneration and sensory improvement in CMT1A mouse models and in patients. Neurology 65:681–689. doi:10.1212/01. wnl.0000171978.70849.c5 201. Godinho MJ, Teh L, Pollett MA et al (2012) Immunohistochemical, ultrastructural and functional analysis of axonal regeneration through peripheral nerve grafts containing Schwann cells expressing BDNF, CNTF or NT3. PLoS ONE 8:e69987–e69987. doi:10. 1371/journal.pone.0069987 202. Gordon T, Sulaiman O, Boyd JG (2003) Experimental strategies to promote functional recovery after peripheral nerve injuries. J Peripher Nerv Syst 8:236–250 203. English AW, Schwartz G, Meador W et al (2007) Electrical stimulation promotes peripheral axon regeneration by enhanced neuronal

Mol Neurobiol

204.

205.

206.

207.

208.

209.

210.

211.

212.

213.

214. 215.

216.

217.

218. 219. 220.

221. 222.

223.

neurotrophin signaling. Dev Neurobiol 67:158–172. doi:10.1002/ dneu.20339 Nadim W, Anderson PN, Turmaine M (1990) The role of Schwann cells and basal lamina tubes in the regeneration of axons through long lengths of freeze-killed nerve grafts. Neuropathol Appl Neurobiol 16:411–421 Krekoski CA, Neubauer D, Zuo J, Muir D (2001) Axonal regeneration into acellular nerve grafts is enhanced by degradation of chondroitin sulfate proteoglycan. J Neurosci 21:6206–6213 Al-Majed AA, Brushart TM, Gordon T (2000) Electrical stimulation accelerates and increases expression of BDNF and trkB mRNA in regenerating rat femoral motoneurons. Eur J Neurosci 12:4381– 4390 Geremia NM, Gordon T, Brushart TM et al (2007) Electrical stimulation promotes sensory neuron regeneration and growthassociated gene expression. Exp Neurol 205:347–359. doi:10. 1016/j.expneurol.2007.01.040 Al-Majed AA, Tam SL, Gordon T (2004) Electrical stimulation accelerates and enhances expression of regeneration-associated genes in regenerating rat femoral motoneurons. Cell Mol Neurobiol 24:379–402 Sabatier MJ, Redmon N, Schwartz G, English AW (2008) Treadmill training promotes axon regeneration in injured peripheral nerves. Exp Neurol 211:489–493. doi:10.1016/j.expneurol.2008.02.013 English AW, Cucoranu D, Mulligan A, Sabatier M (2009) Treadmill training enhances axon regeneration in injured mouse peripheral nerves without increased loss of topographic specificity. J Comp Neurol 517:245–255. doi:10.1002/cne.22149 English AW, Cucoranu D, Mulligan A et al (2011) Neurotrophin-4/5 is implicated in the enhancement of axon regeneration produced by treadmill training following peripheral nerve injury. Eur J Neurosci 33:2265–2271. doi:10.1111/j.1460-9568.2011.07724.x Buck CR, Seburn KL, Cope TC (2000) Neurotrophin expression by spinal motoneurons in adult and developing rats. J Comp Neurol 416:309–318 Frisén J, Verge VM, Fried K et al (1993) Characterization of glial trkB receptors: differential response to injury in the central and peripheral nervous systems. Proc Natl Acad Sci U S A 90:4971– 4975 Gordon T (2009) The role of neurotrophic factors in nerve regeneration. J Neurosurg 26:E3. doi:10.3171/FOC.2009.26.2.E3 Jang S-W, Liu X, Yepes M et al (2010) A selective TrkB agonist with potent neurotrophic activities by 7,8-dihydroxyflavone. Proc Natl Acad Sci U S A 107:2687–2692. doi:10.1073/pnas. 0913572107 Jang S-W, Liu X, Chan CB et al (2010) Deoxygedunin, a natural product with potent neurotrophic activity in mice. PLoS ONE 5: e11528. doi:10.1371/journal.pone.0011528 English AW, Liu K, Nicolini JM, et al. (2013) Small-molecule trkB agonists promote axon regeneration in cut peripheral nerves. Proc Natl Acad Sci USA 1–6. doi: 10.1073/pnas.1303646110 Yaksh TL (1999) Spinal drug delivery. Elsevier Health Sciences Cook MJ (1965) The anatomy of the laboratory mouse. Academic Press Green EL (1962) Quantitative genetics of skeletal variations in the mouse: II. Crosses between four inbred strains (C3H, DBA, C57BL, BALB/c). Genetics 47:1085–1096 Green EL (1941) Genetic and non-genetic factors which influence the type of the skeleton in an inbred strain of mice. Genetics 26:192–222 Hankenson FC, Garzel LM, Fischer DD et al (2008) Evaluation of tail biopsy collection in laboratory mice (Mus musculus): vertebral ossification, DNA quantity, and acute behavioral responses. J Am Assoc Lab Anim Sci JAALAS 47:10–18 Shinohara H (1999) The mouse vertebrae: changes in the morphology of mouse vertebrae exhibit specific patterns over limited numbers of vertebral levels. Okajimas Folia Anat Japonica 76:17–31

224. Sakla FB (1969) Quantitative studies on the postnatal growth of the spinal cord and the vertebral column of the albino mouse. J Comp Neurol 136:237–247. doi:10.1002/cne.901360209 225. Watson C, Paxinos G, Kayalioglu G (2009) The spinal cord. Academic Press 226. Biscoe TJ, Nickels SM, Stirling CA (1982) Numbers and sizes of nerve fibres in mouse spinal roots. Q J Exp Physiol (Cambridge, England) 67:473–494 227. Altman J, Bayer SA (1984) The development of the rat spinal cord. Adv Anat Embryol Cell Biol 85:1–164 228. Rexed B (1952) The cytoarchitectonic organization of the spinal cord in the cat. J Comp Neurol 96:414–495 229. Molander C, Xu Q, Grant G (1984) The cytoarchitectonic organization of the spinal cord in the rat: I. The lower thoracic and lumbosacral cord. J Comp Neurol 230:133–141. doi:10.1002/cne. 902300112 230. Caspary T, Anderson KV (2003) Patterning cell types in the dorsal spinal cord: what the mouse mutants say. Nat Rev Neurosci 4:289– 297. doi:10.1038/nrn1073 231. Todd AJ (2010) Neuronal circuitry for pain processing in the dorsal horn. Nat Rev Neurosci 11:823–836. doi:10.1038/nrn2947 232. Ferri CC, Moore FA, Bisby MA (1998) Effects of facial nerve injury on mouse motoneurons lacking the p75 low-affinity neurotrophin receptor. J Neurobiol 34:1–9 233. Casaccia-Bonnefil P, Carter BD, Dobrowsky RT, Chao MV (1996) Death of oligodendrocytes mediated by the interaction of nerve growth factor with its receptor p75. Nature 383:716–719. doi:10. 1038/383716a0 234. Beattie MS, Harrington AW, Lee R et al (2002) ProNGF induces p75-mediated death of oligodendrocytes following spinal cord injury. Neuron 36:375–386 235. Michael GJ, Kaya E, Averill S et al (1997) TrkA immunoreactive neurones in the rat spinal cord. J Comp Neurol 385:441–455 236. Ramer MS, Bradbury EJ, McMahon SB (2001) Nerve growth factor induces P2X(3) expression in sensory neurons. J Neurochem 77: 864–875. doi:10.1046/j.1471-4159.2001.00288.x 237. Scarisbrick IA, Isackson PJ, Windebank AJ (1999) Differential expression of brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 in the adult rat spinal cord: regulation by the glutamate receptor agonist kainic acid. J Neurosci 19:7757– 7769 2 3 8 . Ya n Q , R a d e k e M J , M a t h e s o n C R e t a l ( 1 9 9 7 ) Immunocytochemical localization of TrkB in the central nervous system of the adult rat. J Comp Neurol 378:135–157 239. Merlio JP, Ernfors P, Jaber M, Persson H (1992) Molecular cloning of rat trkC and distribution of cells expressing messenger RNAs for members of the trk family in the rat central nervous system. Neuroscience 51:513–532 240. Zhou L-J, Yang T, Wei X et al (2011) Brain-derived neurotrophic factor contributes to spinal long-term potentiation and mechanical hypersensitivity by activation of spinal microglia in rat. Brain Behav Immun 25:322–334. doi:10.1016/j.bbi.2010.09.025 241. Thompson SW, Bennett DL, Kerr BJ et al (1999) Brain-derived neurotrophic factor is an endogenous modulator of nociceptive responses in the spinal cord. Proc Natl Acad Sci U S A 96:7714– 7718 242. Miletic G, Miletic V (2002) Increases in the concentration of brain derived neurotrophic factor in the lumbar spinal dorsal horn are associated with pain behavior following chronic constriction injury in rats. Neurosci Lett 319:137–140 243. McMahon SB, Cafferty W (2004) Neurotrophic influences on neuropathic pain. Novartis Found Symp 261:68–102 244. Yajima Y, Narita M, Narita M et al (2002) Involvement of a spinal brain-derived neurotrophic factor/full-length TrkB pathway in the development of nerve injury-induced thermal hyperalgesia in mice. Brain Res Rev 958:338–346

Mol Neurobiol 245. Wang X, Ratnam J, Zou B et al (2009) TrkB signaling is required for both the induction and maintenance of tissue and nerve injuryinduced persistent pain. J Neurosci 29:5508–5515. doi:10.1523/ JNEUROSCI.4288-08.2009 246. Michael GJ, Averill S, Shortland PJ et al (1999) Axotomy results in major changes in BDNF expression by dorsal root ganglion cells: BDNF expression in large trkB and trkC cells, in pericellular baskets, and in projections to deep dorsal horn and dorsal column nuclei. Eur J Neurosci 11:3539–3551 247. Li L, Xian CJ, Zhong J-H, Zhou X-F (2006) Upregulation of brainderived neurotrophic factor in the sensory pathway by selective motor nerve injury in adult rats. Neurotox Res 9:269–283 248. Slack SE, Grist J, Mac Q et al (2005) TrkB expression and phosphoERK activation by brain-derived neurotrophic factor in rat spinothalamic tract neurons. J Comp Neurol 489:59–68. doi:10. 1002/cne.20606 249. Matsumoto T, Rauskolb S, Polack M et al (2008) Biosynthesis and processing of endogenous BDNF: CNS neurons store and secrete BDNF, not pro-BDNF. Nat Neurosci 11:131–133. doi:10.1038/ nn2038 250. Srinivasan B, Roque CH, Hempstead BL et al (2004) Microgliaderived pronerve growth factor promotes photoreceptor cell death via p75 neurotrophin receptor. J Biol Chem 279:41839–41845. doi: 10.1074/jbc.M402872200 251. Ulmann L, Hatcher JP, Hughes JP et al (2008) Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain. J Neurosci 28: 11263–11268. doi:10.1523/JNEUROSCI.2308-08.2008 252. Middlemas DS, Lindberg RA, Hunter T (1991) trkB, a neural receptor protein-tyrosine kinase: evidence for a full-length and two truncated receptors. Mol Cell Biol 11:143–153 253. Biffo S, Offenhauser N, Carter BD, Barde YA (1995) Selective binding and internalisation by truncated receptors restrict the availability of BDNF during development. Development (Cambridge, England) 121:2461–2470 254. Eide FF, Vining ER, Eide BL et al (1996) Naturally occurring truncated trkB receptors have dominant inhibitory effects on brain-derived neurotrophic factor signaling. J Neurosci 16:3123– 3129 255. Saarelainen T, Pussinen R, Koponen E et al (2000) Transgenic mice overexpressing truncated trkB neurotrophin receptors in neurons have impaired long-term spatial memory but normal hippocampal LTP. Synapse (New York, NY) 38:102–104. doi:10.1002/10982396(200010)38:13.0.CO;2-K 256. Luikart BW, Nef S, Shipman T, Parada LF (2003) In vivo role of truncated trkb receptors during sensory ganglion neurogenesis. Neuroscience 117:847–858 257. Dorsey SG, Renn CL, Carim-Todd L et al (2006) In vivo restoration of physiological levels of truncated TrkB.T1 receptor rescues neuronal cell death in a trisomic mouse model. Neuron 51:21–28. doi: 10.1016/j.neuron.2006.06.009 258. Carim-Todd L, Bath KG, Fulgenzi G et al (2009) Endogenous truncated TrkB.T1 receptor regulates neuronal complexity and TrkB kinase receptor function in vivo. J Neurosci 29:678–685. doi:10.1523/JNEUROSCI.5060-08.2009 259. Baxter GT, Radeke MJ, Kuo RC et al (1997) Signal transduction mediated by the truncated trkB receptor isoforms, trkB.T1 and trkB.T2. J Neurosci 17:2683–2690 260. Yacoubian TA, Lo DC (2000) Truncated and full-length TrkB receptors regulate distinct modes of dendritic growth. Nat Neurosci 3:342–349. doi:10.1038/73911 261. Renn CL, Leitch CC, Dorsey SG (2009) In vivo evidence that truncated trkB.T1 participates in nociception. Mol Pain 5:61 262. Rose CR, Blum R, Pichler B et al (2003) Truncated TrkB-T1 mediates neurotrophin-evoked calcium signalling in glia cells. Nature 426:74–78. doi:10.1038/nature01983

263. Dreyfus CF, Dai X, Lercher LD et al (1999) Expression of neurotrophins in the adult spinal cord in vivo. J Neurosci Res 56: 1–7. doi:10.1002/(SICI)1097-4547(19990401)56:13.0.CO;2-3 264. Woolf CJ, Ma Q (2007) Nociceptors—noxious stimulus detectors. Neuron 55:353–364. doi:10.1016/j.neuron.2007.07.016 265. Whiteside GT, Munglani R (2001) Cell death in the superficial dorsal horn in a model of neuropathic pain. J Neurosci Res 64: 168–173 266. Scholz J, Broom DC, Youn D-H et al (2005) Blocking caspase activity prevents transsynaptic neuronal apoptosis and the loss of inhibition in lamina II of the dorsal horn after peripheral nerve injury. J Neurosci 25:7317–7323. doi:10.1523/JNEUROSCI.152605.2005 267. Torsney C, MacDermott AB (2006) Disinhibition opens the gate to pathological pain signaling in superficial neurokinin 1 receptorexpressing neurons in rat spinal cord. J Neurosci 26:1833–1843. doi:10.1523/JNEUROSCI.4584-05.2006 268. Hathway GJ, Vega-Avelaira D, Moss A et al (2009) Brief, low frequency stimulation of rat peripheral C-fibres evokes prolonged microglial-induced central sensitization in adults but not in neonates. Pain 144:110–118. doi:10.1016/j.pain.2009.03.022 269. Suter MR, Berta T, Gao Y-J et al (2009) Large A-fiber activity is required for microglial proliferation and p38 MAPK activation in the spinal cord: different effects of resiniferatoxin and bupivacaine on spinal microglial changes after spared nerve injury. Mol Pain 5: 53. doi:10.1186/1744-8069-5-53 270. Calvo M, Bennett DLH (2012) The mechanisms of microgliosis and pain following peripheral nerve injury. Exp Neurol 234:271–282. doi:10.1016/j.expneurol.2011.08.018 271. Tsuda M, Shigemoto-Mogami Y, Koizumi S et al (2003) P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 424:778–783. doi:10.1038/nature01786 272. Ginhoux F, Greter M, Leboeuf M et al (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science (New York, NY) 330:841–5. doi:10.1126/ science.1194637 273. Ransohoff RM, Perry VH (2009) Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol 27:119–145. doi:10.1146/annurev.immunol.021908.132528 274. Kim SU, de Vellis J (2005) Microglia in health and disease. J Neurosci Res 81:302–313. doi:10.1002/jnr.20562 275. Nimmerjahn A, Kirchhoff F, Helmchen F (2005) Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science (New York, NY) 308:1314–1318. doi:10.1126/science. 1110647 276. White FA, Jung H, Miller RJ (2007) Chemokines and the pathophysiology of neuropathic pain. Proc Natl Acad Sci U S A 14: 20151–20158 277. Beggs S, Trang T, Salter MW (2012) P2X4R(+) microglia drive neuropathic pain. Nat Neurosci 15:1068–1073. doi:10.1038/nn. 3155 278. Martin S, Vincent J-P, Mazella J (2003) Involvement of the neurotensin receptor-3 in the neurotensin-induced migration of human microglia. J Neurosci 23:1198–1205 279. Dicou E, Vincent J-P, Mazella J (2004) Neurotensin receptor-3/ sortilin mediates neurotensin-induced cytokine/chemokine expression in a murine microglial cell line. J Neurosci Res 78:92–99. doi: 10.1002/jnr.20231 280. Eriksson NP, Persson JK, Svensson M et al (1993) A quantitative analysis of the microglial cell reaction in central primary sensory projection territories following peripheral nerve injury in the adult rat. Exp Brain Res 96:19–27 281. Beggs S, Salter MW (2007) Stereological and somatotopic analysis of the spinal microglial response to peripheral nerve injury. Brain Behav Immun 21:624–633. doi:10.1016/j.bbi.2006.10.017

Mol Neurobiol 282. Coyle DE (1998) Partial peripheral nerve injury leads to activation of astroglia and microglia which parallels the development of allodynic behavior. Glia 23:75–83 283. Zhang F, Vadakkan KI, Kim SS et al (2008) Selective activation of microglia in spinal cord but not higher cortical regions following nerve injury in adult mouse. Mol Pain. doi:10.1186/1744-8069-4-15 284. Tsuda M, Kuboyama K, Inoue T et al (2009) Behavioral phenotypes of mice lacking purinergic P2X. Mol Pain. doi:10.1186/1744-80695-28 285. Bennett MVL, Garré JM, Orellana JA et al (2012) Connexin and pannexin hemichannels in inflammatory responses of glia and neurons. Brain Res Rev 1487:3–15. doi:10.1016/j.brainres.2012.08. 042 286. Foley JC, McIver SR, Haydon PG (2011) Gliotransmission modulates baseline mechanical nociception. Mol Pain 7:93. doi:10.1186/ 1744-8069-7-93 287. Zhuang ZY, Wen YR, Zhang DR et al (2006) A Peptide c-Jun NTerminal Kinase (JNK) Inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci 26:3551–3560. doi:10.1523/ JNEUROSCI.5290-05.2006 288. Colburn RW, DeLeo JA, Rickman AJ et al (1997) Dissociation of microglial activation and neuropathic pain behaviors following peripheral nerve injury in the rat. J Neuroimmunol 79:163– 175 289. Nakagawa T, Wakamatsu K, Zhang N et al (2007) Intrathecal administration of ATP produces long-lasting allodynia in rats: differential mechanisms in the phase of the induction and maintenance. Neuroscience 147:445–455. doi:10.1016/j.neuroscience.2007.03.045 290. Liu W, Tang Y, Feng J (2011) Cross talk between activation of microglia and astrocytes in pathological conditions in the central nervous system. Life Sci 89:141–146. doi:10.1016/j.lfs.2011.05.011 291. Bechmann I, Galea I, Perry VH (2007) What is the blood–brain barrier (not)? Trends Immunol 28:5–11. doi:10.1016/j.it.2006.11. 007 292. Sofroniew MV, Vinters HV (2009) Astrocytes: biology and pathology. Acta Neuropathol 119:7–35. doi:10.1007/s00401-009-0619-8 293. Miyoshi K, Obata K, Kondo T et al (2008) Interleukin-18-mediated microglia/astrocyte interaction in the spinal cord enhances neuropathic pain processing after nerve injury. J Neurosci 28:12775– 12787. doi:10.1523/JNEUROSCI.3512-08.2008 294. Jin S-X, Zhuang Z-Y, Woolf CJ, Ji R-R (2003) p38 mitogenactivated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci 23:4017– 4022 295. Piao ZG, Cho I-H, Park CK et al (2006) Activation of glia and microglial p38 MAPK in medullary dorsal horn contributes to tactile hypersensitivity following trigeminal sensory nerve injury. Pain 121:219–231. doi:10.1016/j.pain.2005.12.023 296. Trang T, Beggs S, Wan X, Salter MW (2009) P2X4-receptormediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation. J Neurosci 29:3518–3528. doi:10.1523/ JNEUROSCI.5714-08.2009

297. Gomes C, Ferreira R, George J et al (2013) Activation of microglial cells triggers a release of brain-derived neurotrophic factor (BDNF) inducing their proliferation in an adenosine A2A receptordependent manner: A2A receptor blockade prevents BDNF release and proliferation of microglia. J Neuroinflamm 10:16. doi:10.1038/ nm1103 298. Torsney C, MacDermott AB (2005) Neuroscience: a painful factor. Nature 438:923–925. doi:10.1038/438923a 299. Yang J, Siao C-J, Nagappan G et al (2009) Neuronal release of proBDNF. Nat Neurosci 12:113–115. doi:10.1038/nn.2244 300. Woo NH, Teng HK, Siao CJ et al (2005) Activation of p75NTR by proBDNF facilitates hippocampal long-term depression. Nat Neurosci 8:1069–1077. doi:10.1038/nn1510 301. Petersen CM, Nielsen MS, Nykjaer A et al (1997) Molecular identification of a novel candidate sorting receptor purified from human brain by receptor-associated protein affinity chromatography. J Biol Chem 272:3599–3605 302. Coull JAM, Boudreau D, Bachand K et al (2003) Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain. Nature 424:938–942. doi:10.1038/nature01868 303. Coull JAM, Beggs S, Boudreau D et al (2005) BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 438:1017–1021. doi:10.1038/nature04223 304. Bohlhalter S, Weinmann O, Mohler H, Fritschy JM (1996) Laminar compartmentalization of GABAA-receptor subtypes in the spinal cord: an immunohistochemical study. J Neurosci 16:283–297 305. Bormann J, Hamill OP, Sakmann B (1987) Mechanism of anion permeation through channels gated by glycine and gammaaminobutyric acid in mouse cultured spinal neurones. J Physiol 385:243–286 306. Jonas P, Bischofberger J, Sandkühler J (1998) Corelease of two fast neurotransmitters at a central synapse. Science (New York, NY) 281:419–424 307. Keller AF, Coull JAM, Chéry N et al (2001) Region-specific developmental specialization of GABA–glycine cosynapses in laminas I– II of the rat spinal dorsal horn. J Neurosci 21:7871–7880 308. Rivera C, Voipio J, Thomas-Crusells J et al (2004) Mechanism of activity-dependent downregulation of the neuron-specific K-Cl cotransporter KCC2. J Neurosci 24:4683–4691. doi:10.1523/ JNEUROSCI.5265-03.2004 309. Nabekura J, Ueno T, Okabe A et al (2002) Reduction of KCC2 expression and GABAA receptor-mediated excitation after in vivo axonal injury. J Neurosci 22:4412–4417 310. Rivera C, Li H, Thomas-Crusells J et al (2002) BDNF-induced TrkB activation down-regulates the K+–Cl− cotransporter KCC2 and impairs neuronal Cl- extrusion. J Cell Biol 159:747–752. doi: 10.1083/jcb.200209011 311. Zhang X, Wang J, Zhou Q et al (2011) Brain-derived neurotrophic factor-activated astrocytes produce mechanical allodynia in neuropathic pain. Neuroscience 199:452–460. doi:10.1016/j. neuroscience.2011.10.017 312. Constandil L, Goich M, Hernández A et al (2012) Cyclotraxin-B, a new TrkB antagonist, and glial blockade by propentofylline, equally prevent and reverse cold allodynia induced by BDNF or partial infraorbital nerve constriction in mice. J Pain 13:579–589. doi:10. 1016/j.jpain.2012.03.008

Peripheral nerve injury modulates neurotrophin signaling in the peripheral and central nervous system.

Peripheral nerve injury disrupts the normal functions of sensory and motor neurons by damaging the integrity of axons and Schwann cells. In contrast t...
1MB Sizes 0 Downloads 6 Views