12
REVIEW Downstream effector molecules in successful peripheral nerve regeneration Smriti Patodia & Gennadij Raivich Received: 29 February 2012 / Accepted: 19 March 2012 / Published online: 15 May 2012 # Springer-Verlag 2012 Abstract The robust axon regeneration that occurs follow- ing peripheral nerve injury is driven by transcriptional acti- vation of the regeneration program and by the expression of a wide range of downstream effector molecules from neuro- peptides and neurotrophic factors to adhesion molecules and cytoskeletal adaptor proteins. These regeneration-associated effector molecules regulate the actin-tubulin machinery of growth-cones, integrate intracellular signalling and stimula- tory and inhibitory signals from the local environment and translate them into axon elongation. In addition to the neuro- nally derived molecules, an important transcriptional com- ponent is found in locally activated Schwann cells and macrophages, which release a number of cytokines, growth factors and neurotrophins that support neuronal survival and axonal regeneration and that might provide directional guid- ance cues towards appropriate peripheral targets. This review aims to provide a comprehensive up-to-date account of the transcriptional regulation and functional role of these effector molecules and of the information that they can give us with regard to the organisation of the regeneration program. Keywords Nerve regeneration . Peripheral nerve injury . Downstream effector molecules . Adhesion molecules . Cytoskeletal adaptor proteins Introduction Injury to peripheral nerves sets in motion a targeted program of neurite outgrowth, culminating in the re-innervation of targets (skin, muscle, visceral organs, blood vessels) and recovery of motor, sensory and autonomic function. This functionally successful regeneration depends both on the activation of the intrinsic growth capacity of neurons and on the presence of a permissive environment with axon guidance cues (Chen et al. 2007). This is also clearly emphasised by comparison with the centrally projecting central and primary sensory neurons, where a combination of diminished intrinsic capacity for regeneration and height- ened presence of inhibitory factors of their extracellular environment results in poor regeneration (Buchli et al. 2007; Bolsover et al. 2008; Gordon et al. 2009). Chromatolysis and transcriptional changes Axonal injury generates several major signalling cues to the neuronal cell body (Raivich and Makwana 2007), which undergoes a chromatolyticreaction (Lieberman 1971). This includes the interruption of the normal flow of trophic signals (Raivich et al. 1991; Zigmond and Sun 1997) and the exposure of the transected axons to new signals in the extracellular environment (Sendtner et al. 1997), all of which results in the retrograde transport of de novo activated molecules to the cell body within 12-24 h following injury (Hanz et al. 2003; Ben-Yaakov et al. 2012). Disruption of the tight ionic concentration gradient through membrane leaks also causes the rapid elevation of Ca 2+ and cAMP levels, transmitting successive injury-mediated action S. Patodia : G. Raivich (*) Centre for Perinatal Brain Protection and Repair, University College London, Chenies Mews 86-96, London WC1E 6HX, UK e-mail: [email protected] Cell Tissue Res (2012) 349:1526 DOI 10.1007/s00441-012-1416-6

Downstream Effector Molecules in Successful Peripheral

Embed Size (px)

DESCRIPTION

paper

Citation preview

Page 1: Downstream Effector Molecules in Successful Peripheral

REVIEW

Downstream effector molecules in successful peripheralnerve regeneration

Smriti Patodia & Gennadij Raivich

Received: 29 February 2012 /Accepted: 19 March 2012 /Published online: 15 May 2012# Springer-Verlag 2012

Abstract The robust axon regeneration that occurs follow-ing peripheral nerve injury is driven by transcriptional acti-vation of the regeneration program and by the expression ofa wide range of downstream effector molecules from neuro-peptides and neurotrophic factors to adhesion molecules andcytoskeletal adaptor proteins. These regeneration-associatedeffector molecules regulate the actin-tubulin machinery ofgrowth-cones, integrate intracellular signalling and stimula-tory and inhibitory signals from the local environment andtranslate them into axon elongation. In addition to the neuro-nally derived molecules, an important transcriptional com-ponent is found in locally activated Schwann cells andmacrophages, which release a number of cytokines, growthfactors and neurotrophins that support neuronal survival andaxonal regeneration and that might provide directional guid-ance cues towards appropriate peripheral targets. Thisreview aims to provide a comprehensive up-to-date accountof the transcriptional regulation and functional role of theseeffector molecules and of the information that they can giveus with regard to the organisation of the regenerationprogram.

Keywords Nerve regeneration . Peripheral nerve injury .

Downstream effector molecules . Adhesion molecules .

Cytoskeletal adaptor proteins

Introduction

Injury to peripheral nerves sets in motion a targeted programof neurite outgrowth, culminating in the re-innervation oftargets (skin, muscle, visceral organs, blood vessels) andrecovery of motor, sensory and autonomic function. Thisfunctionally successful regeneration depends both on theactivation of the intrinsic growth capacity of neurons andon the presence of a permissive environment with axonguidance cues (Chen et al. 2007). This is also clearlyemphasised by comparison with the centrally projectingcentral and primary sensory neurons, where a combinationof diminished intrinsic capacity for regeneration and height-ened presence of inhibitory factors of their extracellularenvironment results in poor regeneration (Buchli et al.2007; Bolsover et al. 2008; Gordon et al. 2009).

Chromatolysis and transcriptional changes

Axonal injury generates several major signalling cues to theneuronal cell body (Raivich and Makwana 2007), whichundergoes a “chromatolytic” reaction (Lieberman 1971).This includes the interruption of the normal flow of trophicsignals (Raivich et al. 1991; Zigmond and Sun 1997) andthe exposure of the transected axons to new signals in theextracellular environment (Sendtner et al. 1997), all ofwhich results in the retrograde transport of de novo activatedmolecules to the cell body within 12-24 h following injury(Hanz et al. 2003; Ben-Yaakov et al. 2012). Disruption ofthe tight ionic concentration gradient through membraneleaks also causes the rapid elevation of Ca2+ and cAMPlevels, transmitting successive injury-mediated action

S. Patodia :G. Raivich (*)Centre for Perinatal Brain Protection and Repair,University College London,Chenies Mews 86-96,London WC1E 6HX, UKe-mail: [email protected]

Cell Tissue Res (2012) 349:15–26DOI 10.1007/s00441-012-1416-6

Page 2: Downstream Effector Molecules in Successful Peripheral

potentials, which, in turn, activate a cascade of signallingpathways, including the mitogen-activated protein kinases(extracellular signal-regulated kinase [ERK], p38, c-JunN-terminal kinase [JNK]), Janus kinases (JAK)/signal trans-ducers and activators of transcription (STAT), Ras/Raf,phosphatidylinositol 3 kinase (PI3K)/AKT, abl, mammaliantarget of rapamycin (mTOR) and Mstb3 pathways (Chang etal. 2003; Lindwall and Kanje 2005; Chen et al. 2007;Michaelevski et al. 2010).

This activation of cytoplasmic signalling pathways israpidly followed by the appearance and nuclear transloca-tion of a host of transcription factors such as c-jun, jun D,ATF3 (cyclic AMP-dependent transcription factor), STAT3,P311, p53, CREB (cAMP response element-binding), NFAT(nuclear factor of activated T-cells), KLF (Krüppel-likefactor), Sox11 (sex-determining region Y box-containing 11)and C/EBP (CCAAT/enhancer binding protein) β, δ(Schwaiger et al. 2000; Mason et al. 2003; Raivich et al.2004; Nadeau et al. 2005; Di Giovanni et al. 2006; Jankowskiet al. 2009; Magoulas and Lopez-de Heredia 2010; Moore andGoldberg 2011; Ruff et al. 2012; Patodia and Raivich 2012).Transcription factors provide a vital link between injury-induced signals and downstream protein expression via generegulation. They rapidly condition the injured nerve and,within 1–4 days after injury, the neuronal perikaryon producesa plethora of regeneration associated genes (RAGs), anumbrella term including the vast number of genes that aredifferentially regulated during nerve regeneration and thatmight be involved in cell-cell signalling, axonal growth andsprouting and the activation of the non-neuronal cellularmilieu (Skene and Willard 1981; Verhaagen et al. 1986;Boeshore et al. 2004). A summary of signalling from earlysensors to cytoplasmic mediators to transcription factors andthe synthesis of effector molecules is shown in Fig. 1.

Neuronal deletion of transcription factors has frequentlyshown reduced regenerative ability. For example, the removalof genes encoding c-Jun and STAT3 have been shown tostrongly decrease the speed of axonal regeneration, persistentlyreduce target re-innervation, and delay functional recovery(Patodia et al. 2011; Ruff et al. 2012). A number of differentdownstream effector targets of these transcription factors arealmost completely abolished, as seen in the case of neuro-nal c-jun deletion (Fig. 2). Interestingly, functional recoverytends to catch up after prolonged periods of regeneration,possibly because of rerouting and collateral sprouting,whereas anatomical reinnervation is still brought down; incases of c-Jun and STAT3 deletion, the whisker hair pad, amajor target of facial nerve regeneration, still only receivesapproximately a third of the normal number of axons at3 months after nerve cut (Raivich et al. 2004; Patodia et al.2011). Interestingly, the effects of STAT3 deletion aremilder in the dorsal root ganglia (Bareyre et al. 2011;

Ben-Yaakov et al. 2012), suggesting the presence of com-plementary pathways, alongside STAT3, in the peripheralsensory neurons.

Proregenerative effects have also been shown for p53(Di Giovanni et al. 2006) and, to a lesser extent, forSox11 and C/EBPδ (Jankowski et al. 2009; Magoulasand Lopez-de Heredia 2010). In the case of STAT3,strong pro-regenerative effects have also been observedwhen the upstream regulation of this transcription factoris modified. Inactivation of floxed neuronal SOCS3, anupstream inhibitor of STAT3, by using viral vector car-rying cre recombinase elicits pronounced axonal regener-ation in the central crushed optic nerve model (Sun et al.2011). This effect can be blocked by the concurrentdeletion of floxed STAT3. In a similar vein, deletion ofgp130, the common neurokine receptor subunit thattransduces the effects of interleukin-6 (IL6), leukemiainhibitory factor (LIF), ciliary neurotrophic factor(CNTF), etc. in the sympathetic neurons of the superiorcervical ganglion abolishes up-regulation, phosphorylationand nuclear translocation of STAT3 after postgangionicaxotomy, while also blocking most of the injury response(Hyatt Sachs et al. 2010).

Although N-terminal phosphorylation of c-Jun plays animportant role in modifying transcriptional activity and elic-its strong effects in stroke, ischaemia and excitotoxicty, itseffects on regeneration are less clear. Unlike complete neu-ronal deletion, with a strikingly persistent defect in targetreinnervation and neuronal cell death (Raivich et al. 2004),the deletion of the more upstream Jun N-terminal kinasesonly shows a moderate transient effect on functional recov-ery (Ruff et al. 2012). Likewise, the replacement of theserine 63 and 73 and the threonine 91 and 93 phosphoac-ceptor residues with alanines only reproduces some of theatrophy and delayed functional recovery (S. Patodia, A.Behrens and G. Raivich, unpublished; Ruff et al. 2012).However, upstream signals leading to c-Jun chemical mod-ification are not confined to the N-terminus: glycogen-synthase-kinase (GSK) phosphorylation of threonine 239,activity that is inhibited by ERK and PI3K signals, enhancesJun ubiquitination and degradation (Morton et al. 2003; Weiet al. 2005) and thus, in the presence of ERK or PI3Kactivation, Jun would become more stable. Activatingeffects on Jun function have also been observed with thep300 acetylation of lysine 268 and 271 (Vries et al. 2001;Wang et al 2006). Either of these or an additional upstreamsignal-mediated modification might play a role in Junregeneration signalling but this will need to be demonstratedin vivo by selective amino-acid replacements, similar to theexchange of the N-terminal serines and threonines.

Overall, because transcription factors are able to bind topromoters of many different target genes, they can switch

16 Cell Tissue Res (2012) 349:15–26

Page 3: Downstream Effector Molecules in Successful Peripheral

on/off the expression of a diverse range of downstreameffector molecules. An overview of the transcriptional reg-ulation of the expression and function of downstream effec-tor molecules in peripheral nerve regeneration is shown inTable 1. In this review, we will focus on the roles andregulatory mechanisms of such identified downstream effec-tors in successful peripheral nerve regeneration, specificallyaxonal adhesion molecules, chemoattractant signaling, cellsurface-cytoskeletal adaptors and neuropeptides but alsoneurotrophins produced in the injured nerve. Here, recentadvances in cre/loxP technology permit cell-type and/ortime-specific genetic knockouts (Sauer 1998; Akira 2000)and have begun to provide useful insights into theirupstream transcriptional regulation and downstream func-tion in orchestrating complex axon growth and regenerativeresponses.

Adhesion molecules

Adhesion molecules enable the interaction of the cell surfaceof the axonal growth conewith the adjacent bands of Büngner,which consist in denervated Schwann cells and with the innerlining of the neural tube basal membrane; both structures actas a scaffold for the growing axon (Grumet 1991; Shapiro et

al. 2007). Signaling between adhesion molecules on axonaland Schwann cell surfaces is frequently bi-directional(Quintes et al. 2010; Fricker and Bennett 2011) but, in orderto simplify this for the purpose of axonal regeneration, it ishelpful to concentrate on axonal molecules that act as recep-tors for axonal guidance cues from Schwann cells and theassociated extracellular matrix.

Regenerating neurons up-regulate a variety of adhesionmolecules. These include integrins that can serve as receptorsfor the matrix and cell surface molecules such as laminin,fibronectin and paxillin (Kloss et al. 1999; Werner et al.2000; Vogelezang et al. 2001, 2007; Ekstrom et al. 2003;Wallquist et al. 2004; Gardiner et al. 2005). The expressionof CD44, a receptor for hyaluronic acid (Jones et al. 2000) andof galectin-1, a lectin receptor for the galactoside side chains(Horie and Kadoya 2000; Akazawa et al. 2004), increases.Regenerating axons express cell multimodal surface mole-cules involved in homophilic binding such as ninjurin (nerveinjury-induced protein; Araki and Milbrandt 2000), gicerin/CD146, a heterophilic receptor for the neurite outgrowthfactor (Taira et al. 1994) and adhesion molecule FLTR3,which can form complexes with receptors for fibroblastgrowth factor (FGF) and potentiate FGF signalling (Bottcheret al. 2004). On the functional side, administration of exoge-nous galectin-1 promotes neurite outgrowth, whereas its

Fig. 1 Cascade of cellularsignalling in successfulregeneration, from early sensorsof axonal injury, to cytoplasmicsignals, transcriptionalactivation and downstreameffectors. Modified from Figure4 in Raivich (2011)

Cell Tissue Res (2012) 349:15–26 17

Page 4: Downstream Effector Molecules in Successful Peripheral

removal by antibody neutralisation or through gene deletiondecreases the speed of neurite outgrowth (Horie and Kadoya2000; McGraw et al. 2004). Removal of β2-microglobulinproduces a moderate 20 % reduction in the ability of regener-ating motor axons to cross the proximal to distal gap afternerve cut (Oliveira et al. 2004).

Finally, regenerating axons also contain an assortment ofdegradative enzymes including urokinase and plasminogenactivator (PA), metalloproteinases (MMP) 2, 3 and 9(Demestre et al. 2004; Shubayev and Myers 2004) anddamage-induced neuronal endopeptidase (DINE; Kiryu-Seoet al. 2000). Activation of MMP2 and MMP9 is partiallydependent on the presence of tissue PA (tPA) and/or urokinasePA (UPA) systems (Siconolfi and Seeds 2003) and the dele-tion of tPA and/or UPA interferes with functional recoveryafter peripheral nerve injury (Siconolfi and Seeds 2001). Aswith chondroitinase (Graham et al. 2007; Hattori et al. 2008;Udina et al. 2010), the administration of exogenous MMP2has been shown to degrade chondroitin sulphate proteogly-cans and to improve recovery and anatomical reinnervationfollowing nerve transection; furthermore, in the presence ofsaturating concentrations of MMP2, the co-administration of

chondrotinase ABC does not produce any additional improve-ment, suggesting that both enzymes attack the same target(Zuo et al. 1998a, 1998b). Interestingly, although regeneratingsympathetic neurons show an increase in MMP2 proteinactivity, no comparative increase occurs in MMP2 mRNA(Leone et al. 2005). However, the axotomised dorsal rootganglion neurons show injury-caused down-regulation ofTIMP2, an inhibitor of MMP2 (Huang et al. 2011) and asimilar downward trend is seen in the facial motor nucleus(G. Raivich, M.R. Mason, J. Verhaagen, unpublished), sug-gesting a regeneration-induced disinhibition of MMP2.

So far, three from the above group of adhesion moleculesup-regulated after injury, namely DINE (Kiryu-Seo et al.2008), CD44 (Jones et al. 2000) and the α7β1 integrin(Werner et al. 2000), have also been identified as in vivodownstream targets of the regeneration-associated transcrip-tion factors mentioned in the above section. Deletion ofneuronal c-Jun or STAT3 interferes with the up-regulationof CD44 and of the α7 and β1 integrin subunits (Raivich etal. 2004; Patodia et al. 2011; Ruff et al. 2012). Preliminaryin vivo data show the same regulatory pattern for DINE (S.Kiryu-Seo, H. Kiyama, G. Raivich, unpublished); in vitro,

Fig. 2 Summary of selecteddownstream effector targets ofneuronal c-Jun. Overviewshowing immunohistochemistryfor the facial nucleus at 14 daysafter nerve cut in control mice(jun f/f; a, b, e, f, i, j, m, n, q, r)and in junΔS mutant (syn junf7F; c, d, g, h, k, l, o, p, s, t) micein which the floxed c-Jun genewas deleted with Cre under thecontrol of the synapsin promoter.Columns 1, 3 Contralateral side(co). Columns 2, 4 Axotomisedside (ax). After axotomy, jun f/fmice show a prominent increasein adhesion molecules CD44(a-d) and β1 integrin subunit(e-h), in neuropeptides calcitoningene-related peptide (CGRP; i-l)and galanin (Galn; m-p) and intranscription factor ATF3 (q-t);these changes are reduced orabolished in the junΔS mutants.Bar 0.2 mm. Reproduced fromFigure 5 in Ruff et al. (2012)

18 Cell Tissue Res (2012) 349:15–26

Page 5: Downstream Effector Molecules in Successful Peripheral

Table 1 Overview of the transcriptional regulation of expression andfunction of downstream effector molecules in successful peripheral nerveregeneration (STAT signal transducer and activator of transcription 3,

ATF3 cyclic AMP-dependent transcription factor, N neuronal, SCSchwann cell, NMJ neuromuscular junction, ↑ increase)

Downstreameffectors (DE)

Transcriptionfactors driving DE

Expressionafter axotomy

Effects of DE deletion on nerveregeneration

References

Adhesion molecules

α7β1 Integrin c-jun (N), STAT3(N)

↑ Global α7 deletion strongly reducesregenerative speed after nerve crush

Werner et al. 2000

Raivich et al. 2004

Similar effects with neuronal β1 deletion Patodia et al. 2011

S. Patodia, C. Santos and G.Raivich, unpublished

CD44 c-jun (N), STAT3(N)

↑ Reduced neurite outgrowth oftransplanted central noradrenergicneurons

Nagy et al. 1998

Lin and Chan 2003

Errors in retinal axonal growth indevelopment

Raivich et al. 2004

Patodia et al. 2011However, no data regarding effectson regeneration, as yet Ruff et al. 2012

DINE (damage-inducedneuronalendopeptidase)

ATF3, c-jun,STAT3

↑ Reduced sprouting at developing NMJ Kiryu-Seo et al. 2008

However, no data regarding effects onregeneration, as yet

Nagata et al. 2010

Neuropeptides

Galanin c-jun (N), STAT3(N)

↑ Reduced rate of regeneration Holmes et al. 2000

Modulation of pain transmission afterinjury

Raivich et al. 2004

Holmes et al. 2005

Patodia et al. 2011

Ruff et al. 2012

CGRP (calcitoningene-related peptide)

c-jun (N), STAT3(N)

↑ Reduced number of axons crossingfrom proximal to distal stump

Raivich et al. 2004

Toth et al. 2009

Patodia et al. 2011

Ruff et al. 2012

Guidance molecules

Ephrin-B Sox-2 (SC) ↑ Disordered axonal outgrowth fromproximal to distal stump

Parrinello et al. 2010

Cytoskeletal adaptors

CAP23 (cytoskeleton-associated protein 23)

c-jun (N) ↑ Reduced neurite outgrowth and targetre-innervation

Anderson et al. 2006;

J. Verhaagen, Y. Mattson andG. Raivich, unpublished

GAP43 (neuromodulin) C/EBPβ (N) ↑ Abnormal developmental path-finding Strittmatter et al. 1995

However, no data regarding effects onregeneration, as yet

Nadeau et al. 2005

Neurotrophins

BDNF (brain-derivedneurotrophic factor)

c-jun (SC) ↑ No data regarding effects onregeneration, as yet

Meyer et al. 1992

GDNF (glial-cell-derivedneurotrophic factor)

c-jun (SC) ↑ Increased neuronal cell death X. Fontana et al., in preparationReduced functional recovery and speedof regeneration

Artemin c-jun (SC) ↑ Increased neuronal cell death X. Fontana et al., in preparationReduced functional recovery and speedof regeneration

LIF (leukemiainhibitory factor)

c-jun (SC) ↑ No data regarding effects onregeneration, as yet

Curtis et al. 1994

NGF (nerve growthfactor)

c-fos (SC) ↑ No data regarding effects on regeneration,as yet

Hengerer et al. 1990

Cell Tissue Res (2012) 349:15–26 19

Page 6: Downstream Effector Molecules in Successful Peripheral

DINE expression in neuronal cell cultures is strongly up-regulated by STAT3, in combination with an ATF3/c-Junfusion hybrid (Kiryu-Seo et al. 2008). The data regardingthe neuronal functions of DINE and CD44 come from invitro, explantation and developmental studies. Thus, anti-body inhibition of CD44 reduces neurite outgrowth of trans-planted central noradrenergic neurons (Nagy et al. 1998)and creates multiple errors in retinal axonal growth trajec-tory through the optic chiasm (Lin and Chan 2003). In thecase of DINE, the global deletion of its function following aninsertion of loxP sites into its gene is associatedwith amassivereduction in terminal sprouting of motor axons at the neuro-muscular junction (Nagata et al. 2010). Unfortunately, thesefloxed DINE animals coincidentally suffer from perinatallethality, precluding the direct examination of DINE effect inthe adult by using the Cre/lox system.

In the case of the α7β1 integrin, global deletion of the α7subunit causes a strong reduction of approximately 40 % inthe speed of adult motor axon regeneration in the facialnerve model, with a commensurate delay in the reinnerva-tion of its main peripheral target, namely the whisker pad(Werner et al. 2000). Similarly, deletion of the α7 integrinsubunit also abolishes the ex vivo conditioning effect; theexplantation of axotomised wild-type sensory ganglia invitro leads to brisk neurite outgrowth on laminin, whereasthe outgrowth is weaker and delayed when using previouslyuninjured ganglia (Ekstrom et al. 2003).

Overall, the integrins are a large family of heterodimerictransmembrane glycoproteins composed of specificallypaired α and β subunits. In addition to cell adhesion,migration and axonal outgrowth, integrin signalling isimportant for non-neuronal and neuronal survival (Previtaliet al. 2001; Chen et al. 2007; Lemons and Condic 2008;Tucker and Mearow 2008). Although some α subunits canonly dimerise with single β subunits (α1, α2, α3, α5, α7and α8 only with β1; αL, αD, αM and αX only with β2;αIIb only with β3), others such as α4 (β1, β7), α6 (β1,β4), α9 (β1, β8) and αV (β1, β3, β5, β6, β8) can partnermultiple β subunits (Previtali et al. 2001). Many of the β1-pairing α subunits (α1, α4, α7, α9, etc.) have been shownto promote axonal regeneration in vitro and followingforced expression in vivo (Toyota et al. 1990; Vogelezanget al. 2001; Snider et al. 2002; Andrews et al. 2009), raisingthe expectation that the complete removal of β1 will pro-duce a more severe phenotype. However, preliminary resultsinvolving the neuron-specific deletion of β1 in the facialnerve model suggest that the phenotype is roughly on parwith that observed for the global α7 deletion (S. Patodia, X.Santos and G. Raivich, unpublished). This coincides withthe finding that α7 is the main neuronal α subunit thatappears up-regulated following axotomy (Kloss et al.1999; Werner et al. 2000; Vogelezang et al. 2001; Andrewset al. 2009).

Neuropeptides

Axon transection frequently causes increased expression fora variety of neuropeptides. For example, a pronouncedincrease of calcitonin gene-related peptide (CGRP), galaninand to a lesser extent pituitary adenylate cyclase activatingpeptide (PACAP) occurs in subpopulations of transectedcranial and spinal motoneurons (Moore 1989; Raivich etal. 1995). Sensory dorsal root ganglion neurons show anup-regulation of galanin, vasculointestinal peptide (VIP)and neuropeptide Y (NPY; Xu et al. 1990; Noguchi et al.1993); sympathetic neurons demonstrate an increase in VIP,galanin and substance P. Not every peptide is up-regulatedin every group of neurons: some, such as substance P orCGRP in the axotomised sensory dorsal root ganglia orNPY in the sympathetic neurons, actually show decreasedlevels (Dumoulin et al. 1991; Habecker et al. 2009), similarto the down-regulation of enzymes and transporter systemsfor adrenergic and cholinergic neurotransmitters in the sym-pathetic and motor neurons (Zigmond and Sun 1997; Kallaet al. 2001). Recent studies point to decreased expression ofthe Hand2 transcription factor causing the post-axotomydown-regulation for components of the adrenergic system(Pellegrino et al. 2011); similar pathways might thus beinvolved in the down-regulation of some neuropeptides ininjured neurons.

Several of these up-regulated neuropeptides have alsobeen recently identified as in vivo downstream targets ofthe regeneration-associated transcription factors. Deletion ofneuronal c-Jun or STAT3 interferes with the up-regulation ofCGRP and galanin in facial motoneurons (Raivich et al.2004; Patodia et al. 2011; Ruff et al. 2012). In sympatheticneurons, targeted deletion of the core neurokine receptorgp130, which transduces signals for IL6, LIF, CNTF andother neurokines, show drastically lowered post-axotomyexpression for VIP, galanin and PACAP but does not affectthe up-regulation of cholecystokinin (Habecker et al. 2009).Deletion of gp130 also abolishes the nuclear appearance ofphosphorylated STAT3, suggesting that this transcription factoris also involved in the normally occurring post-axotomyup-regulation of galanin and associated neuropeptides.

At the functional level, the deletion of galanin or its type2 receptor expressed in neurons results in a 35 % reductionin rate of peripheral nerve regeneration after sciatic nervecrush (Holmes et al. 2000) and modulates pain transmission(Holmes et al. 2005). In the case of CGRP, the local inhibi-tion of intra-axonal CGRP synthesis with short interferingRNA (siRNA) strongly reduces the number of axons grow-ing in the conduit between proximal and distal nerve stumps(Toth et al. 2009). Similar inhibitory effects have also beenobserved by blocking the CGRP receptor expressed onneighbouring Schwann cells, via siRNA against the CGRPreceptor activity modifying protein-1, suggesting that the

20 Cell Tissue Res (2012) 349:15–26

Page 7: Downstream Effector Molecules in Successful Peripheral

locally expressed peptide is used to recruit Schwann cellcooperation for neurite outgrowth (Raivich et al. 1992; Tothet al. 2009). In the case of PACAP, homozygous deletion ofthe gene reduces initial neurite outgrowth in the first 24 hafter injury, with a moderate retarding effect (1-2 days) onthe reinnervation of the peripheral target (Armstrong et al.2008). Reverse experiments, with local application of PACAPand galanin, have shown that either peptide will stronglyaugment the peripheral branching of regenerating axons,increasing the number of neurons regenerating into each ofthe identified rami of the facial nerve by up to five-fold(Suarez et al. 2006). However, these pro-sprouting effectsare actually associated with an impaired outcome, under-scoring the importance of misrouting as a major impedi-ment to the resumption of coordinated functional activity.

Guidance signalling

Guidance signals (through cell surface contact or diffusiblesignals mediating attraction or repulsion) play an importantrole by binding to receptors on growth cone surfaces, trig-gering secondary signals and steering axon extensions in thecorrect direction (Ide 1996; Giger et al. 2010). Overall,many different families of guidance molecules have beenfound, including the semaphorins and their receptors; theneuropilins and plexins (Tessier-Lavigne and Goodman1996; Huber et al. 2003), ephrins (Tessier-Lavigne 2002;Koeberle and Bähr 2004; Parrinello et al. 2010), slits(Koeberle and Bähr 2004) or inhibitory myelin componentssuch as MAG (myelin-associated glycoprotein) can activateRho via p75 neurotrophin receptor (p75NTR; Yamashitaet al. 2002).

Functionally, the deletion of neuropilin-2 reduces thedensity of the light neurofilament-positive axons in the distalnerve and interferes with functional recovery (Lindholm etal. 2004; Bannerman et al. 2008). Axotomy induces theexpression of Bex1, an intracellular adaptor molecule thatinteracts with p75NTR to reduce neurite outgrowth inhibi-tion by myelin inhibitors (Lindholm et al. 2004). Globaldeletion of Bex1 reduces the number of regenerating axonscrossing the sciatic nerve crush site by >50 % and impedesfunctional recovery (Khazaei et al. 2010). Interestingly, thedeletion of p75 does not affect neuronal survival or the speedof axonal regeneration in the facial nerve model, although p75null mice show an enhanced neuroinflammatory responsecompared with their wild-type littermates (Gschwendtneret al. 2003).

In most cases, the in vivo transcriptional regulation forthese molecules in neurons has not been clearly delineated.However, some of the Schwann cell function in creatingaxonal guidance scaffolds is under the control of post-traumatically expressed transcription factor Sox-2, induced

by fibroblast ephrin-B signalling. Acting on the Schwanncell EphB2-receptor, the fibroblast ligand induces Sox2 and,in a Sox2-dependent manner, N-cadherin required for sort-ing Schwann cells into cell cords that link proximal anddistal nerve stump (Parrinello et al. 2010). Interference withthis process through EphB2 deletion or blocking antibodiesproduces a significantly more disordered axonal outgrowthin the gap region between the proximal and distal part ofthe nerve.

Growth cone and cytoskeletal adaptors

Appearing at the tip of growing axons, the growth cones arespecialised quasi-autonomous structures that are responsiblefor growth, path-finding and recognition of targets afternerve injury. In agreement with their navigatory function,they are heavily decorated with adhesion molecules andreceptors for chemoattractive and repulsive signals, as dis-cussed above. Inside the growth cone, cytoskeletal adaptorsplay a crucial role in mediating connections between the cellsurface and cytoskeletal actin-microtubule core of the grow-ing axons (Baas and Ahmad 2001; Ellezam et al. 2002;Zhang et al. 2003; Bouquet et al. 2004; Madura et al. 2004).

One important family of adaptor molecules is the “GMC”family, which includes “integral” membrane proteins, namelyGAP43/neuromodulin, myristoylated alanine-rich C kinasesubstrate (MARCKS) and cytoskeleton-associated protein 23(CAP23; Skene andWillard 1981; Verhaagen et al. 1986; Freyet al. 2000; Bomze et al. 2001). GAP43 and CAP23 areamong the most abundant proteins in axonal growth cones(Goslin and Banker 1990; Bomze et al. 2001). These proteinsco-distribute with phosphoinositol-4, 5-diphosphate (PIP2) atthe semi-crystalline plasmalemmal raft regions and modifyraft-recruitment of signalling molecules such as src (Laux etal. 2000), bind to acidic phospholipids such as PIP2, calcium/calmodulin, protein kinase C and actin filaments in a mutuallyexclusive manner, alter actin cytoskeleton polymerisation,organisation and disassembly and translate receptor-mediatedcalcium fluxes into signals guiding growth cone activity (Skene1990; Ide 1996; Laux et al. 2000; Henley and Poo 2004;Kulbatski et al. 2004). In vitro, the depletion of GAP43 doesnot impair nerve growth factor (NGF)-elicited neurite out-growth but leads to poorer adhesion, unstable lamellar exten-sions devoid of local F-actin, reduced branching and enhancedsensitivity to inhibitory stimuli (Aigner and Caroni 1995). Invivo, the global deletion of GAP43 interferes with normaldevelopmental pathfinding (Strittmatter et al. 1995) and thatof CAP23 with inactivity-induced sprouting at the neuromus-cular synapse (Frey et al. 2000). Overexpression of GAP43 orCAP23 induces excessive neuromuscular sprouting (Caroni etal. 1997) and the combined overexpression of both adaptorcomponents (GAP43 and CAP23) strongly enhances neurite

Cell Tissue Res (2012) 349:15–26 21

Page 8: Downstream Effector Molecules in Successful Peripheral

growth from peripheral sensory neurons into the injuredadult mouse spinal cord, with little regeneration being observedif only one of the two components is overexpressed (Bomzeet al. 2001).

After axotomy, the transcriptional up-regulation of themRNAs encoding GAP43 and CAP23 is biphasic and isdown-regulated after target reconnection (Mason et al. 2002).In the case of GAP43, the early phase (24 h after axotomy) isc/EBPβ-independent but is followed by a c/EBPβ-dependentlater phase, with an almost complete disappearance ofincreased GAP43 mRNA in c/EBPβ null mutants at day 3(Nadeau et al. 2005). In the case of CAP23, up-regulation in thefirst 24 h is unaffected but the second phase of 4-14 days isreduced in the absence of neuronal c-Jun (J. Verhaagen, Y.Mattson and G. Raivich, unpublished observations). Since thelong-term postnatal survival of GAP43 null mice is rare(Strittmatter et al. 1995; but see Donovan et al. 2002),studies in the adult have been limited to the CAP23deletion (Caroni et al. 1997; Frey et al. 2000). Recent dataobtained by using the neuron-specific deletion of the floxedCAP23 gene with the adenoassociated virus carrying Cre-recombinase (Anderson et al. 2006) or by selective neuro-nal expression of Cre under the synapsin promoter syn::cre(S. Patodia and P.N. Anderson, unpublished) reveal anapproximately 50 % reduction in the reinnervation of thewhisker pad, the main target of regenerating facial nervefibers (Werner et al. 2000).

Neurotrophins, growth factors and cytokines

Peripheral nerve injury causes a massive increase in thesynthesis and/or availability of a variety of neurotrophicand growth-promoting factors (Frostick et al. 1998; Raivichand Makwana 2007). These include neurotrophin-3 (NT3;Terenghi 1999) and NT4/5 (English et al. 2005), NGF(Heumann et al. 1987), brain-derived neurotrophic factor(BDNF; Meyer et al. 1992), glial-cell-derived neurotrophicfactor (GDNF; Naveilhan et al. 1997), insulin-like growthfactors-1/2 (IGF1, IGF2; Kanje et al. 1989; Glazner et al.1993), basic FGF (Jungnickel et al. 2004), vascular endo-thelial growth factor (Islamov et al. 2004), LIF (Curtis et al.1994; Haas et al. 1999), CNTF (Kirsch et al. 2003),interleukin-1 (IL1; Lindholm et al. 1988), IL6 (Murphy etal. 1995; Hirota et al. 1996) and transforming growth factor-β1 (Lindholm et al. 1992).

On the functional side, exogenous application of IGF1and IGF2 enhances the pinch-test-determined speed of axo-nal regeneration, whereas their antibody-mediated inhibitionreduces this speed (Kanje et al. 1989; Glazner et al. 1993);IGF1 is also partly responsible for peripheral conditioning(Kanje et al. 1991). Peripheral nerve grafts from mice lack-ing NT4/5 display decreased ingrowth by regenerating

axons of wild-type animals (English et al. 2005). Theabsence of the Schwann cell CNTF in CNTF null mice alsoappears to delay the appearance of phosphorylated STAT3and its nuclear translocation in neuronal cell bodies (Kirschet al. 2003).

Studies of the transcriptional regulation of induced neu-rotrophin and growth factor expression in peripheral nervehave concentrated particularly on the role of c-Fos andc-Jun. Sciatic nerve lesions cause a rapid local increase inc-fos and c-jun mRNA followed, within hours, by a peri-trauma increase in NGF mRNA (Hengerer et al. 1990), withendoneurial fibroblasts forming a primary site of NGF syn-thesis (Heumann et al. 1987; Lindholm et al. 1988). Thatthese two sets of changes, namely the up-regulation of c-Fosand the later expression of NGF, are probably linked hasbeen shown by an elegant set of cell culture experiments.Here, heavy-metal-induced overexpression of c-Fos infibroblasts from transgenic mice with metallothionein-promoter-driven Fos results in a rapid up-regulation ofc-Jun and NGF, with the c-Fos/Jun heterodimer binding tothe AP1 site in the first intron of the NGF gene, an eventthat is critical for NGF mRNA transcription (Hengereret al. 1990).

Recent studies have also underscored the importance ofthe c-Jun counterpart. Here, the Schwann-cell-specific dele-tion of floxed c-Jun by using p0 promoter driven Cre (p0::cre) produces a massive increase in facial motoneuron celldeath, with a commensurate reduction in peripheral targetreinnervation and in functional recovery (X. Fontana et al.,in preparation). Follow-on analysis of neurotrophin andgrowth factor expression in the injured sciatic nerves ofp0::cre jun flox/flox mice (abbreviated as p0:jun) hasrevealed a strong deficit in many of the normally up-regulated trophic factors, including GDNF, Artemin, BDNFand LIF. Interestingly, the administration of recombinantGDNF and Artemin to these p0:jun mice substantiallyreduces post-traumatic neuronal cell death and improvesfunctional recovery, underscoring the importance ofSchwann cell c-Jun in providing trophic support for injuredand regenerating neurons.

Concluding remarks

The current summary provides an overview of the sequenceof events starting from injury signals via transcription fac-tors and the appearance of downstream effectors involved indriving axonal regeneration and functional recovery.Amongst these effectors, adhesion and guidance molecules,cytoskeletal adaptors, neuropeptides and trophic factors allappear to be tangibly involved. In many cases, the directinjury-associated transcriptional regulation is still unclear;for example, little is known about the factors involved in the

22 Cell Tissue Res (2012) 349:15–26

Page 9: Downstream Effector Molecules in Successful Peripheral

down-regulation of the sensitivity to inhibitory and chemo-repulsive stimuli in peripheral regeneration, even thoughevidence exists for the presence of such regeneration blockers.Nevertheless, recent advances obtained by using cell-type-specific deletion of transcription factors enable more and morepieces of the puzzle to be joined together, making visiblebroad lines of an identified chain of activities contributing torecovery and underscoring the role of these transcriptionfactors as master-switches of the regeneration program.

References

Aigner L, Caroni P (1995) Absence of persistent spreading, branching,and adhesion in GAP-43-depleted growth cones. J Cell Biol128:647–660

Akazawa C, Nakamura Y, Sango K, Horie H, Kohsaka S (2004)Distribution of the galectin-1 mRNA in the rat nervous system:its transient upregulation in rat facial motor neurons after facialnerve axotomy. Neuroscience 125:171–178

Akira S (2000) Roles of STAT3 defined by tissue-specific gene targeting.Oncogene 19:2607–2611

Anderson PN, Zhang JY, Ahmed BY, Sablitsky F, Raivich G, LiebermanAR, Verhaagen J (2006) Deletion of CAP-23 in adult mouse motorneurons delays axonal regeneration following facial nerve transec-tion. Soc Neurosci Abstr 522.27/C46

Andrews MR, Czvitkovich S, Dassie E, Vogelaar CF, Faissner A, BlitsB, Gage FH, ffrench-Constant C, Fawcett JW (2009) Alpha9integrin promotes neurite outgrowth on tenascin-C and enhancessensory axon regeneration. J Neurosci 29:5546–5557

Araki T, Milbrandt J (2000) Ninjurin2, a novel homophilic adhesionmolecule, is expressed in mature sensory and enteric neurons andpromotes neurite outgrowth. J Neurosci 20:187–195

Armstrong BD, Abad C, Chhith S, Cheling-Lau G, Hajji OE, NoblitaH, Waschek JA (2008) Impaired nerve regeneration and enhancedneuroinflammatory response in mice lacking pituitary adenylylcyclase activating peptide. Neuroscience 151:63–73

Baas PW, Ahmad FJ (2001) Force generation by cytoskeletal motorproteins as a regulator of axonal elongation and retraction. TrendsCell Biol 11:244–249

Bannerman P, Ara J, Hahn A, Hong L, McCauley E, Friesen K,Pleasure D (2008) Peripheral nerve regeneration is delayed inneuropilin 2-deficient mice. J Neurosci Res 86:3163–3169

Bareyre FM, Garzorz N, Lang C, Misgeld T, Büning H, KerschensteinerM (2011) In vivo imaging reveals a phase-specific role of STAT3during central and peripheral nervous system axon regeneration.Proc Natl Acad Sci USA 108:6282–6287

Ben-Yaakov K, Dagan SY, Segal-Ruder Y, Shalem O, Vuppalanchi D,Willis DE, Yudin D, Rishal I, Rother F, Bader M, Blesch A, PilpelY, Twiss JL, Fainzilber M (2012) Axonal transcription factorssignal retrogradely in lesioned peripheral nerve. EMBO J31:1350–1363

Boeshore KL, Schreiber RC, Vaccariello SA, Sachs HH, Salazar R,Lee J, Ratan RR, Leahy P, Zigmond RE (2004) Novel changes ingene expression following axotomy of a sympathetic ganglion: amicroarray analysis. J Neurobiol 59:216–235

Bolsover S, Fabes J, Anderson PN (2008) Axonal guidance moleculesand the failure of axonal regeneration in the adult mammalianspinal cord. Restor Neurol Neurosci 26:117–130

Bomze HM, Bulsara KR, Iskandar BJ, Caroni P, Skene JHP (2001)Spinal axon regeneration evoked by replacing two growth coneproteins in adult neurons. Nat Neurosci 4:38–43

Böttcher RT, Pollet N, Delius H, Niehrs C (2004) The transmembraneprotein XFLRT3 forms a complex with FGF receptors and pro-motes FGF signalling. Nat Cell Biol 6:38–44

Bouquet C, Soares S, Boxberg Y von, Ravaille-Veron M, Propst F,Nothias F (2004) Microtubule-associated protein 1B controlsdirectionality of growth cone migration and axonal branching inregeneration of adult dorsal root ganglia neurons. J Neurosci24:7204–7213

Buchli AD, Rouiller E, Mueller R, Dietz V, Schwab ME (2007) Repairof the injured spinal cord. A joint approach of basic and clinicalresearch. Neurodegener Dis 4:51–56

Caroni P, Aigner L, Schneider C (1997) Intrinsic neuronal determinantslocally regulate extrasynaptic and synaptic growth at the adultneuromuscular junction. J Cell Biol 136:679–692

Chang F, Steelman LS, Lee JT, Shelton JG, Navolanic PM, BlalockWL, Franklin RA, McCubrey JA (2003) Signal transductionmediated by the Ras/Raf/MEK/ERK pathway from cytokinereceptors to transcription factors: potential targeting for therapeuticintervention. Leukemia 17:1263–1293

Chen ZL, Yu WM, Strickland S (2007) Peripheral regeneration. AnnuRev Neurosci 30:209–233

Curtis R, Scherer SS, Somogyi R, Adryan KM, Ip NY, Zhu Y, LindsayRM, DiStefano PS (1994) Retrograde axonal transport of LIF isincreased by peripheral nerve injury: correlation with increasedLIF expression in distal nerve. Neuron 12:191–204

Demestre M, Wells GM, Miller KM, Smith KJ, Hughes RA, GearingAJ, Gregson NA (2004) Characterisation of matrix metalloprotei-nases and the effects of a broad-spectrum inhibitor (BB-1101) inperipheral nerve regeneration. Neuroscience 124:767–779

Di Giovanni S, Knights CD, Rao M, Yakovlev A, Beers J, Catania J,Avantaggiati ML, Faden AI (2006) The tumor suppressor proteinp53 is required for neurite outgrowth and axon regeneration.EMBO J 25:4084–4096

Donovan SL, Mamounas LA, Andrews AM, Blue ME, McCasland JS(2002) GAP-43 is critical for normal development of the seroto-nergic innervation in forebrain. J Neurosci 22:3543–3552

Dumoulin FL, Raivich G, Streit WJ, Kreutzberg GW (1991) Differentialregulation of calcitonin gene-related peptide (CGRP) in regeneratingrat facial nucleus and dorsal root ganglion. Eur J Neurosci 3:338–342

Ekström PA, Mayer U, Panjwani A, Pountney D, Pizzey J, Tonge DA(2003) Involvement of alpha7beta1 integrin in the conditioning-lesion effect on sensory axon regeneration. Mol Cell Neurosci22:383–395

Ellezam B, Dubreuil C, Winton M, Loy L, Dergham P, Sellés-Navarro I,McKerracher L (2002) Inactivation of intracellular Rho to stimulateaxon growth and regeneration. Prog Brain Res 137:371–380

English AW, Meador W, Carrasco DI (2005) Neurotrophin-4/5 isrequired for the early growth of regenerating axons in peripheralnerves. Eur J Neurosci 21:2624–2634

Frey D, Laux T, Xu L, Schneider C, Caroni P (2000) Shared andunique roles of CAP23 and GAP43 in actin regulation, neuriteoutgrowth, and anatomical plasticity. J Cell Biol 149:1443–1453

Fricker FR, Bennett DL (2011) The role of neuregulin-1 in the responseto nerve injury. Future Neurol 6:809–822

Frostick SP, Yin Q, Kemp GJ (1998) Schwann cells, neurotrophicfactors, and peripheral nerve regeneration. Microsurgery18:397–405

Gardiner NJ, Fernyhough P, Tomlinson DR, Mayer U, Von der MarkH, Streuli CH (2005) Alpha 7 integrin mediates neurite outgrowthof distinct populations of adult sensory neurons. Mol Cell Neurosci28:229–240

Giger RJ, Hollis ER, Tuszynski MH (2010) Guidance molecules inaxon regeneration. Cold Spring Harb Perspect Biol 2:a001867

Glazner GW, Lupien S, Miller JA, Ishii DN (1993) Insulin-like growthfactor-II increases the rate of sciatic-nerve regeneration in rats.Neuroscience 54:791–797

Cell Tissue Res (2012) 349:15–26 23

Page 10: Downstream Effector Molecules in Successful Peripheral

Gordon T, Chan KM, Sulaiman OA, Udina E, Amirjani N, BrushartTM (2009) Accelerating axon growth to overcome limitations infunctional recovery after peripheral nerve injury. Neurosurgery 65(4 Suppl):A132–A144

Goslin K, Banker G (1990) Rapid changes in the distribution ofGAP-43 correlate with the expression of neuronal polarityduring normal development and under experimental conditions.J Cell Biol 110:1319–1331

Graham JB, Neubauer D, Xue QS, Muir D (2007) Chondroitinaseapplied to peripheral nerve repair averts retrograde axonal regen-eration. Exp Neurol 203:185–195

Grumet M (1991) Cell adhesion molecules and their subgroups in thenervous system. Curr Opin Neurobiol 1:370–376

Gschwendtner A, Liu Z, Hucho T, Bohatschek M, Kalla R, Dechant G,Raivich G (2003) Regulation, cellular localization, and functionof the p75 neurotrophin receptor (p75NTR) during the regenera-tion of facial motoneurons. Mol Cell Neurosci 24:307–322

Haas CA, Hofmann HD, Kirsch M (1999) Expression of CNTF/LIF-receptor components and activation of STAT3 signaling in axo-tomized facial motoneurons: evidence for a sequential postle-sional function of the cytokines. J Neurobiol 41:559–571

Habecker BA, Sachs HH, Rohrer H, Zigmond RE (2009) The depen-dence on gp130 cytokines of axotomy induced neuropeptideexpression in adult sympathetic neurons. Dev Neurobiol69:392–400

Hanz S, Perlson E, Willis D, Zheng JQ, Massarwa R, Huerta JJ,Koltzenburg M, Kohler M, van-Minnen J, Twiss JL, FainzilberM (2003) Axoplasmic importins enable retrograde injury signal-ing in lesioned nerve. Neuron 40:1095–1104

Hattori T, Matsuyama Y, Sakai Y, Ishiguro N, Hirata H, Nakamura R(2008) Chondroitinase ABC enhances axonal regeneration acrossnerve gaps. J Clin Neurosci 15:185–191

Hengerer B, Lindholm D, Heumann R, Rüther U, Wagner EF, ThoenenH (1990) Lesion-induced increase in nerve growth factor mRNAis mediated by c-fos. Proc Natl Acad Sci USA 87:3899–3903

Henley J, Poo M (2004) Guiding neuronal growth cones using Ca2+

signals. Trends Cell Biol 14:320–330Heumann R, Lindholm D, Bandtlow C, Meyer M, Radeke MJ, Misko

TP, Shooter E, Thoenen H (1987) Differential regulation ofmRNA encoding nerve growth factor and its receptor in rat sciaticnerve during development, degeneration, and regeneration: role ofmacrophages. Proc Natl Acad Sci USA 84:8735–8739

Hirota H, Kiyama H, Kishimoto T, Taga T (1996) Accelerated nerveregeneration in mice by upregulated expression of interleukin (IL)6 and IL-6 receptor after trauma. J Exp Med 183:2627–2634

Holmes FE, Mahoney S, King VR, Bacon A, Kerr NC, Pachnis V,Curtis R, Priestley JV, Wynick D (2000) Targeted disruption ofthe galanin gene reduces the number of sensory neurons and theirregenerative capacity. Proc Natl Acad Sci USA 97:11563–11568

Holmes FE, Mahoney SA, Wynick D (2005) Use of genetically engi-neered transgenic mice to investigate the role of galanin in theperipheral nervous system after injury. Neuropeptides 39:191–199

Horie H, Kadoya T (2000) Identification of oxidized galectin-1 as aninitial repair regulatory factor after axotomy in peripheral nerves.Neurosci Res 38:131–137

Huang B, Zhao X, Zheng LB, Zhang L, Ni B, Wang YW (2011)Different expression of tissue inhibitor of metalloproteinase familymembers in rat dorsal root ganglia and their changes after peripheralnerve injury. Neuroscience 193:421–428

Huber AB, Kolodkin AL, Ginty DD, Cloutier JF (2003) Signaling atthe growth cone: ligand-receptor complexes and the control ofaxon growth and guidance. Annu Rev Neurosci 26:509–563

Hyatt Sachs H, Rohrer H, Zigmond RE (2010) The conditioning lesioneffect on sympathetic neurite outgrowth is dependent on gp130cytokines. Exp Neurol 223:516–522

Ide C (1996) Peripheral nerve regeneration. Neurosci Res 25:101–121

Islamov RR, Chintalgattu V, Pak ES, Katwa LC, Murashov AK (2004)Induction of VEGF and its Flt-1 receptor after sciatic nerve crushinjury. Neuroreport 15:2117–2121

Jankowski MP, McIlwrath SL, Jing XT, Cornuet PK, Salerno KM,Koerber HR, Albers KM (2009) Sox11 transcription factor mod-ulates peripheral nerve regeneration in adult mice. Brain Res1256:43–54

Jones LL, Liu Z, Shen J, Werner A, Kreutzberg GW, Raivich G (2000)Regulation of the cell adhesionmolecule CD44 after nerve transectionand direct trauma to the mouse brain. J Comp Neurol 426:468–492

Jungnickel J, Gransalke K, Timmer M, Grothe C (2004) Fibroblastgrowth factor receptor 3 signaling regulates injury-related effectsin the peripheral nervous system. Mol Cell Neurosci 25:21–29

Kalla R, Liu Z, Xu S, Koppius A, Imai Y, Kloss CU, Kohsaka S,Gschwendtner A,Moller JC,Werner A, Raivich G (2001)Microgliaand the early phase of immune surveillance in the axotomized facialmotor nucleus: impaired microglial activation and lymphocyterecruitment but no effect on neuronal survival or axonal regenerationin macrophage-colony stimulating factor-deficient mice. J CompNeurol 436:182–201

Kanje M, Skottner A, Sjoberg J, Lundborg G (1989) Insulin-Likegrowth factor-I (Igf-I) stimulates regeneration of the rat sciatic-nerve. Brain Res 486:396–398

Kanje M, Skottner A, Lundborg G, Sjöberg J (1991) Does insulin-likegrowth factor I (IGF-1) trigger the cell body reaction in the ratsciatic nerve? Brain Res 563:285–287

Khazaei MR, Halfter H, Karimzadeh F, Koo JH, Margolis FL, Young P(2010) Bex1 is involved in the regeneration of axons after injury. JNeurochem 115:910–920

Kirsch M, Terheggen U, Hofmann HD (2003) Ciliary neurotrophicfactor is an early lesion-induced retrograde signal for axotomizedfacial motoneurons. Mol Cell Neurosci 24:130–138

Kiryu-Seo S, Sasaki M, Yokohama H, Nakagomi S, Hirayama T, AokiS, Wada K, Kiyama H (2000) Damage-induced neuronal endo-peptidase (DINE) is a unique metallopeptidase expressed in re-sponse to neuronal damage and activates superoxide scavengers.Proc Natl Acad Sci USA 97:4345–4350

Kiryu-Seo S, Kato R, Ogawa T, Nakagomi S, Nagata K, Kiyama H(2008) Neuronal injury-inducible gene is synergistically regulatedby ATF3, c-Jun, and STAT3 through the interaction with Sp1 indamaged neurons. J Biol Chem 283:6988–6996

Kloss CU,Werner A, KleinMA, Shen J, Menuz K, Probst JC, KreutzbergGW, Raivich G (1999) Integrin family of cell adhesion molecules inthe injured brain: regulation and cellular localization in the normaland regenerating mouse facial motor nucleus. J Comp Neurol411:162–178

Koeberle PD, Bähr M (2004) Growth and guidance cues for regeneratingaxons: where have they gone? J Neurobiol 59:162–180

Kulbatski I, Cook DJ, Tator CH (2004) Calcium entry through L-typecalcium channels is essential for neurite regeneration in culturedsympathetic neurons. J Neurotrauma 21:357–374

Laux T, Fukami K, Thelen M, Golub T, Frey D, Caroni P (2000)GAP43, MARCKS, and CAP23 modulate PI(4,5)P(2) at plasma-lemmal rafts, and regulate cell cortex actin dynamics through acommon mechanism. J Cell Biol 149:1455–1472

Lemons ML, Condic ML (2008) Integrin signaling is integral toregeneration. Exp Neurol 209:343–352

Leone L, De Stefano ME, Del Signore A, Petrucci TC, Paggi P (2005)Axotomy of sympathetic neurons activates the metalloproteinase-2 enzymatic pathway. J Neuropathol Exp Neurol 64:1007–1017

Lieberman AR (1971) The axon reaction: a review of the principalfeatures of perikaryal responses to axon injury. Int Rev Neurobiol14:49–124

Lin L, Chan SO (2003) Perturbation of CD44 function affects chias-matic routing of retinal axons in brain slice preparations of themouse retinofugal pathway. Eur J Neurosci 17:2299–2312

24 Cell Tissue Res (2012) 349:15–26

Page 11: Downstream Effector Molecules in Successful Peripheral

Lindholm D, Heumann R, Hengerer B, Thoenen H (1988) Interleukin1 increases stability and transcription of mRNA encoding nervegrowth factor in cultured rat fibroblasts. J Biol Chem 263:16348–16351

Lindholm D, Castrén E, Kiefer R, Zafra F, Thoenen H (1992) Trans-forming growth factor-beta 1 in the rat brain: increase after injuryand inhibition of astrocyte proliferation. J Cell Biol 117:395–400

Lindholm T, Sköld MK, Suneson A, Carlstedt T, Cullheim S, RislingM (2004) Semaphorin and neuropilin expression in motoneuronsafter intraspinal motoneuron axotomy. Neuroreport 15:649–654

Lindwall C, Kanje M (2005) Retrograde axonal transport of JNKsignaling molecules influence injury induced nuclear changes inp-c-Jun and ATF3 in adult rat sensory neurons. Mol Cell Neurosci29:269–282

Madura T, Yamashita T, Kubo T, Tsuji L, Hosokawa K, Tohyama M(2004) Changes in mRNA of Slit-Robo GTPase-activating protein2 following facial nerve transection. Brain Res Mol Brain Res123:76–80

Magoulas B, Lopez-de Heredia L (2010) Functional analysis of thetranscription factor C/EBPdelta in neuronal repair. FENS Abstr5:040.12

Mason MR, Lieberman AR, Grenningloh G, Anderson PN (2002)Transcriptional upregulation of SCG10 and CAP-23 is correlatedwith regeneration of the axons of peripheral and central neurons invivo. Mol Cell Neurosci 20:595–615

Mason MR, Lieberman AR, Anderson PN (2003) Corticospinal neuronsup-regulate a range of growth-associated genes following intracortical,but not spinal, axotomy. Eur J Neurosci 18:789–802

McGraw J, McPhail LT, Oschipok LW, Horie H, Poirier F, Steeves JD,Ramer MS, Tetzlaff W (2004) Galectin-1 in regenerating moto-neurons. Eur J Neurosci 20:2872–2880

Meyer M, Matsuoka I, Wetmore C, Olson L, Thoenen H (1992)Enhanced synthesis of brain-derived neurotrophic factor in thelesioned peripheral nerve: different mechanisms are responsible forthe regulation of BDNF and NGF mRNA. J Cell Biol 119:45–54

Michaelevski I, Segal-Ruder Y, Rozenbaum M, Medzihradszky KF,Shalem O, Coppola G, Horn-Saban S, Ben-Yaakov K, Dagan SY,Rishal I, Geschwind DH, Pilpel Y, Burlingame AL, Fainzilber M(2010) Signaling to transcription networks in the neuronal retro-grade injury response. Sci Signal 3:ra53

Moore DL, Goldberg JL (2011) Multiple transcription factor familiesregulate axon growth and regeneration. Dev Neurobiol 71:1186–1211

Moore RY (1989) Cranial motor neurons contain either galanin- orcalcitonin gene-related peptide-like immunoreactivity. J CompNeurol 282:512–522

Morton S, Davis RJ, McLaren A, Cohen P (2003) A reinvestigation ofthe multisite phosphorylation of the transcription factor c-Jun.EMBO J 22:3876–3886

Murphy PG, Grondin J, Altares M, Richardson PM (1995) Induction ofinterleukin-6 in axotomized sensory neurons. J Neurosci15:5130–5138

Nadeau S, Hein P, Fernandes KJ, Peterson AC, Miller FD (2005) Atranscriptional role for C/EBP beta in the neuronal response toaxonal injury. Mol Cell Neurosci 29:525–535

Nagata K, Kiryu-Seo S, Maeda M, Yoshida K, Morita T, Kiyama H(2010) Damage-induced neuronal endopeptidase is critical forpresynaptic formation of neuromuscular junctions. J Neurosci30:6954–6962

Nagy JI, Price ML, Staines WA, Lynn BD, Granholm AC (1998) Thehyaluronan receptor RHAMM in noradrenergic fibers contributesto axon growth capacity of locus coeruleus neurons in an intra-ocular transplant model. Neuroscience 86:241–255

Naveilhan P, ElShamy WM, Ernfors P (1997) Differential regulation ofmRNAs for GDNF and its receptors Ret and GDNFR alpha aftersciatic nerve lesion in the mouse. Eur J Neurosci 9:1450–1460

Noguchi K, De León M, Nahin RL, Senba E, Ruda MA (1993)Quantification of axotomy-induced alteration of neuropeptidemRNAs in dorsal root ganglion neurons with special referenceto neuropeptide Y mRNA and the effects of neonatal capsaicintreatment. J Neurosci Res 35:54–66

Oliveira AL, Thams S, Lidman O, Piehl F, Hokfelt T, Karre K, LindaH, Cullheim S (2004) A role for MHC class I molecules insynaptic plasticity and regeneration of neurons after axotomy.Proc Natl Acad Sci USA 101:17843–17848

Parrinello S, Napoli I, Ribeiro S, Digby PW, Fedorova M, ParkinsonDB, Doddrell RD, Nakayama M, Adams RH, Lloyd AC (2010)EphB signaling directs peripheral nerve regeneration throughSox2-dependent Schwann cell sorting. Cell 143:145–155

Patodia S, Raivich G (2012) Role of transcription factors in peripheralnerve regeneration. Front Mol Neurosci 5:8

Patodia S, Hristova M, Makwana M, Rumajogee P, Akira S, Raivich G(2011) Transcription factor STAT3 is required for efficient regen-eration after peripheral nerve injury. Soc Neurosci Abstr 784.01/BB22

PellegrinoMJ, Parrish DC, Zigmond RE, Habecker BA (2011) Cytokinesinhibit norepinephrine transporter expression by decreasing Hand2.Mol Cell Neurosci 46:671–680

Previtali SC, Feltri ML, Archelos JJ, Quattrini A, Wrabetz L, HartungH (2001) Role of integrins in the peripheral nervous system. ProgNeurobiol 64:35–49

Quintes S, Goebbels S, Saher G, Schwab MH, Nave KA (2010)Neuron-glia signaling and the protection of axon function bySchwann cells. J Peripher Nerv Syst 15:10–16

Raivich G (2011) Transcribing the path to neurological recovery—from early signals through transcription factors to downstreameffectors of successful regeneration. Ann Anat 193:248–258

Raivich G, Makwana M (2007) The making of successful axonalregeneration: genes, molecules and signal transduction pathways.Brain Res Rev 53:287–311

Raivich G, Hellweg R, Kreutzberg GW (1991) NGF receptor-mediatedreduction in axonal NGF uptake and retrograde transport followingsciatic nerve injury and during regeneration. Neuron 7:151–164

Raivich G, Dumoulin FL, Streit WJ, Kreutzberg GW (1992) Calcitoningene-related peptide (CGRP) in the regenerating rat sciatic nerve.Restor Neurol Neurosci 4:107-115

Raivich G, Reddington M, Haas CA, Kreutzberg GW (1995) Peptidesin motoneurons. Prog Brain Res 104:3–20

Raivich G, Bohatschek M, Da Costa C, Iwata O, Galiano M, HristovaM, Nateri AS, Makwana M, Riera-Sans L, Wolfer DP, Lipp HP,Aguzzi A, Wagner EF, Behrens A (2004) The AP-1 transcriptionfactor c-Jun is required for efficient axonal regeneration. Neuron43:57–67

Ruff C, Staak N, Patodia S, Kaswich M, Rocha-Ferreira E, Da Costa C,Brecht S, Makwana M, Fontana X, Hristova M, Rumajogee P,Galiano M, Bohatschek M, Herdegen T, Behrens A, Raivich G(2012) Neuronal c-Jun is required for successful axonal regener-ation, but the effects of phosphorylation of its N-terminus aremoderate. J Neurochem (In press)

Sauer B (1998) Inducible gene targeting in mice using the Cre/loxsystem. Methods 14:381–392

Schwaiger FW,Hager G, Schmitt AB, Horvat A, Hager G, Streif R, SpitzerC, Gamal S, Breuer S, Brook GA, Nacimiento W, Kreutzberg GW(2000) Peripheral but not central axotomy induces changes in Januskinases (JAK) and signal transducers and activators of transcription(STAT). Eur J Neurosci 12:1165–1176

Sendtner M, Götz R, Holtmann B, Thoenen H (1997) Endogenousciliary neurotrophic factor is a lesion factor for axotomized moto-neurons in adult mice. J Neurosci 17:6999–7006

Shapiro L, Love J, Colman DR (2007) Adhesion molecules in thenervous system: structural insights into function and diversity.Annu Rev Neurosci 30:451–474

Cell Tissue Res (2012) 349:15–26 25

Page 12: Downstream Effector Molecules in Successful Peripheral

Shubayev VI, Myers RR (2004) Matrix metalloproteinase-9 promotesnerve growth factor-induced neurite elongation but not newsprout formation in vitro. J Neurosci Res 77:229–239

Siconolfi LB, Seeds NW (2001) Mice lacking tPA, uPA, or plasminogengenes showed delayed functional recovery after sciatic nerve crush.J Neurosci 21:4348–4355

Siconolfi LB, Seeds NW (2003) Mice lacking tissue plasminogenactivator and urokinase plasminogen activator genes show atten-uated matrix metalloproteases activity after sciatic nerve crush. JNeurosci Res 74:430–434

Skene JH (1990) GAP-43 as a “calmodulin sponge” and some impli-cations for calcium signalling in axon terminals. Neurosci ResSuppl 13:112–125

Skene JHP, Willard M (1981) Characteristics of growth-associatedpolypeptides in regenerating toad retinal ganglion-cell axons. JNeurosci 1:419–426

Snider WD, Zhou FQ, Zhong J, Markus A (2002) Signaling thepathway to regeneration. Neuron 35:13–16

Strittmatter SM, Fankhauser C, Huang PL, Mashimo H, Fishman MC(1995) Neuronal pathfinding is abnormal in mice lacking theneuronal growth cone protein GAP-43. Cell 80:445–452

Suarez V, Guntinas-Lichius O, Streppel M, Ingorokva S, GroshevaM, Neiss WF, Angelov DN, Klimaschewski L (2006) Theaxotomy-induced neuropeptides galanin and pituitary adeny-late cyclase-activating peptide promote axonal sprouting ofprimary afferent and cranial motor neurones. Eur J Neurosci24:1555–1564

Sun F, Park KK, Belin S, Wang D, Lu T, Chen G, Zhang K, Yeung C,Feng G, Yankner BA, He Z (2011) Sustained axon regenerationinduced by co-deletion of PTEN and SOCS3. Nature 480:372–375

Taira E, Takaha N, Taniura H, Kim CH, Miki N (1994) Molecularcloning and functional expression of gicerin, a novel cell adhesionmolecule that binds to neurite outgrowth factor. Neuron 12:861–872

Terenghi G (1999) Peripheral nerve regeneration and neurotrophicfactors. J Anat 194:1–14

Tessier-Lavigne M (2002) Wiring the brain: the logic and molecularmechanisms of axon guidance and regeneration. Harvey Lect98:103–143

Tessier-Lavigne M, Goodman C (1996) The molecular biology of axonguidance. Science 274:1123–1133

Toth CC, Willis D, Twiss JL, Walsh S, Martinez JA, Liu WQ, Midha R,Zochodne DW (2009)Locally synthesized calcitonin gene-relatedpeptide has a critical role in peripheral nerve regeneration. JNeuropathol Exp Neurol 68:326–337

Toyota B, Carbonetto S, David S (1990) A dual laminin/collagenreceptor acts in peripheral nerve regeneration. Proc Natl AcadSci USA 87:1319–1322

Tucker BA, Mearow KM (2008) Peripheral sensory axon growth: fromreceptor binding to cellular signaling. J Neurol Sci 35:551–566

Udina E, Ladak A, Furey M, Brushart T, Tyreman N, Gordon T (2010)Rolipram-induced elevation of cAMP or chondroitinase ABC

breakdown of inhibitory proteoglycans in the extracellular matrixpromotes peripheral nerve regeneration. Exp Neurol 223:143–152

Verhaagen J, Hooff CO van, Edwards PM, De Graan PN, OestreicherAB, Schotman P, Jennekens FG, Gispen WH (1986) The kinase Csubstrate protein B-50 and axonal regeneration. Brain Res Bull17:737–741

Vogelezang MG, Liu Z, Relvas JB, Raivich G, Scherer SS, ffrench-Constant C (2001) Alpha4 integrin is expressed during peripheralnerve regeneration and enhances neurite outgrowth. J Neurosci21:6732–6744

Vogelezang M, Forster UB, Han J, Ginsberg MH, ffrench-Constant C(2007) Neurite outgrowth on a fibronectin isoform expressedduring peripheral nerve regeneration is mediated by the interac-tion of paxillin with alpha4beta1 integrins. BMC Neurosci 8:44

Vries RGJ, Prudenziati M, Zwartjes C, Verlaan M, Kalkhoven E,Zantema A (2001) A specific lysine in c-Jun is required fortranscriptional repression by E1A and is acetylated by p300.EMBO J 20:6095–6103

Wallquist W, Zelano J, Plantman S, Kaufman SJ, Cullheim S,Hammarberg H (2004) Dorsal root ganglion neurons up-regulatethe expression of laminin-associated integrins after peripheral butnot central axotomy. J Comp Neurol 480:162–169

Wang YN, Chen YJ, Chang WC (2006) Activation of extracellularsignal-regulated kinase signaling by epidermal growth factormediates c-jun activation and p300 recruitment in keratin geneexpression. Mol Pharmacol 69:85–98

Wei W, Jin J, Schlisio S, Harper JW, Kaelin WG Jr (2005) The v-Junpoint mutation allows c-Jun to escape GSK3-dependent recognitionand destruction by the Fbw7 ubiquitin ligase. Cancer Cell 8:25–33

Werner A, Willem M, Jones LL, Kreutzberg GW, Mayer U, Raivich G(2000) Impaired axonal regeneration in alpha7 integrin-deficientmice. J Neurosci 20:1822–1830

Xu XJ, Wiesenfeld-Hallin Z, Villar MJ, Fahrenkrug J, Hökfelt T(1990) On the role of galanin, substance P and other neuropep-tides in primary sensory neurons of the rat: studies on spinal reflexexcitability and peripheral axotomy. Eur J Neurosci 2:733–743

Yamashita T, Higuchi H, Tohyama M (2002) The p75 receptor trans-duces the signal from myelin-associated glycoprotein to Rho. JCell Biol 157:565–570

Zhang XF, Schaefer AW, Burnette DT, Schoonderwoert VT, Forscher P(2003) Rho-dependent contractile responses in the neuronalgrowth cone are independent of classical peripheral retrogradeactin flow. Neuron 40:931–944

Zigmond RE, Sun Y (1997) Regulation of neuropeptide expression insympathetic neurons. Paracrine and retrograde influences. Ann NYAcad Sci 814:181–197

Zuo J, Ferguson TA, Hernandez YJ, Stetler-Stevenson WG, Muir D(1998a) Neuronal matrix metalloproteinase-2 degrades and inacti-vates a neurite-inhibiting chondroitin sulfate proteoglycan. J Neurosci18:5203–5211

Zuo J, Hernandez YJ, Muir D (1998b) Chondroitin sulfate proteogly-can with neurite-inhibiting activity is up-regulated followingperipheral nerve injury. J Neurobiol 34:41–54

26 Cell Tissue Res (2012) 349:15–26