Báo cáo khoa học: Molecular mechanisms in successful peripheral regeneration pot

11 461 0
Báo cáo khoa học: Molecular mechanisms in successful peripheral regeneration pot

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

REVIEW ARTICLE Molecular mechanisms in successful peripheral regeneration Milan Makwana 1 and Gennadij Raivich 1,2 1 Centre for Perinatal Brain Protection & Repair, Department of Obstetrics and Gynaecology, University College London, UK 2 Department of Anatomy, University College London, UK Injury to neurons results in a sequence of molecular and cellular responses that are associated with, and that may play an important role, in the mounting of a successful regenerative response, and the ensuing recovery of func- tion. In the injured neuron, the rapid arrival of signals that contribute to cellular injury and stress is followed by the induction of transcription factors, adhesion mole- cules, growth-associated proteins and structural compo- nents needed for axonal elongation. These changes are accompanied by immense shifts in cellular organization: the appearance of growth cones at the proximal tip of the lesioned axons, the swelling of the neuronal cell body associated with a strong increase in cellular metabolism and protein synthesis, and the increase and regional dispersion of areas of rough endoplasmic reti- culum or Nissl shoals in neuronal cytoplasm. This neuronal response is also associated with the expression of growth factors, cytokines, neuropeptides and other secreted molecules involved in cell-to-cell communication, which may be involved in the activa- tion of neighbouring non-neuronal cells around the cell body of the injured neuron and in the distal nerve fibre tracts. In the adult mammalian nervous system peri- pheral nerves show vigorous regeneration. Conversely, axons injured inside central nerve tracts, normally fail to regenerate, regardless of whether their cell bodies are located inside the CNS. Deciphering the molecular and cellular basis for this failure of central regeneration and Keywords adhesion molecule, axonal growth cone, cell survival, cytokine, cytoskeleton, mitogen- activated protein kinase, nerve regeneration, neurotrophin, reinnervation, transcription factor Correspondence G. Raivich, Centre for Perinatal Brain Protection & Repair, Department of Obstetrics & Gynaecology, Department of Anatomy, University College London, 86–96 Chenies Mews, London WC1E 6HX, UK Fax: +44 207 383 7429 Tel: +44 207 679 6068 E-mail: g.raivich@ucl.ac.uk (Received 7 February 2005, accepted 4 April 2005) doi:10.1111/j.1742-4658.2005.04699.x Peripheral nerve injury is normally followed by a robust regenerative response. Here we describe the early changes associated with injury from the initial rise in intracellular calcium and the subsequent activation of transcription factors and cytokines leading to an inflammatory reaction, and the expression of growth factors, cytokines, neuropeptides, and other secreted molecules involved in cell-to-cell communication promoting regen- eration and neurite outgrowth. The aim of this review is to summarize the molecular mechanisms that play a part in executing successful regener- ation. Abbreviations CH1L, close homologue of L1; CNTF, ciliary neurotrophic factor; FGF, fibroblast growth factor; IL, interleukin; MAP, mitogen-associated protein; MAP1B, microtubule-associated protein 1b; MCSF, macrophage colony-stimulating factor; MEK, mitogen-associated protein kinase kinase; NGF, nerve growth factor; NLS, nuclear localization signal; NT3, neurotrophin-3; PI3K, phosphatidyl-inositol-3 kinase; STAT3, signal transducer and activator of transcription-3; TNFR, tumour necrosis factor receptor. 2628 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS strategies to overcome this inhibition is the focus of many research groups and is covered by a number of recent overviews [1–5]. The aim of the current review will be to focus on peripheral nerve regeneration and to describe the molecular and cellular changes that are pivotal in producing a successful regenerative response. Early responses Axonal injury to peripheral nerves normally results in regeneration, although the exact physiological and molecular signals that act as sensors of axonal injury and induce the regenerative programme are debatable. Generally, three major changes appear to provide sep- arate signals. Axonal injury, firstly, interferes with the retrograde flow of signals from the normal innervation target [6,7]; secondly, the tip of the injured axon is exposed to the intracellular content of neighbouring axons and Schwann cells [8,9], and later to the extra- cellular environment of the inflamed neural tissue [10]; and, finally, it causes a rapid entry of extracellular ions such as calcium and sodium through the transiently opened plasma membrane, before it is resealed [11], resulting in depolarization and a sequence of injury- induced action potentials [12]. The calcium influx and bystander activation of intra-axonal proteases and cytoskeleton remodelling underlie growth cone forma- tion [13], and may also be involved in changing intra- axonal protein synthesis [14]. Although the three sets of signals can act in complementary and possibly synergistic roles [15], it remains speculative which of the three, or whether all three are involved in the post- traumatic generation and ⁄ or activation of axoplasmic proteins with the nuclear localization signal (NLS) sequences. These axoplasmic NLS proteins perform an important function as positive injury signals [16,17]. Importantly, peripheral axons and non-neuronal cells contain numerous growth inducing and trophic mole- cules including the ciliary neurotrophic factor (CNTF), neurotrophin-3 (NT3) and fibroblast growth factors (FGFs) [18–20], which are secreted following injury. Both CNTF and the nuclear transcription factor signal transducer and activator of transcription-3 (STAT3), the downstream target of activated CNTF receptor, have regulatory roles that are particularly informative. Neuronal deletion of STAT3 results in enhanced post- traumatic cell death, implicating a neurotrophic-like role for STAT3 [21]. Deletion of CNTF, which is abundantly present in myelinated Schwann cells but not in or around the cell bodies of axotomized motoneurons [22,23] causes a delay in the appearance of the phos- phorylated STAT3 and its nuclear translocation in neuronal cell bodies. Therefore, in summary, this points to a signalling cascade beginning with local release of CNTF by damaged myelinated Schwann cells, its local action on adjacent axons, intra-axonal phosphorylation of STAT3 and its retrograde transport to the cell bodies of injured neurons. At later stages, axonal injury also leads to the recruitment of leukocytes, the production of inflammatory cytokines and attendant changes in neighbouring non-neuronal cells with the synthesis of neurotrophins, chemokines, extracellular matrix mole- cules and proteolytic enzymes (reviewed in [24–26]) changing the extracellular milieu of the lesioned axons. Nerve injury also interferes with the retrograde flow of signals from the normal innervation target, which may lead to the emergence of a negative, denervation signal, following the disconnection. Data on nerve growth factor (NGF), a neurotrophin that is retro- gradely transported from periphery to neuronal cell body are particularly informative. Analysis of its retro- grade transport in the injured sciatic nerve shows a drastic, almost 10-fold decrease immediately following axotomy [6] and although this strong decrease is only short-lived, for approximately 48 h, a three-fold reduc- tion continues until the onset of regeneration. Further- more, NGF deprivation by neutralizing the endogenous activity with specific antibodies induces axotomy-like changes even in the intact, NGF-sensitive sensory and sympathetic neurons, particularly with expression of transcription factors such as c-jun and numerous neuropeptides including galanin, vasoactive intestinal peptide (VIP), substance P (SP), calcitonin gene related peptide (CGRP) and neuropeptide Y (NPY) [7,27]. Aside from these profound biochemical changes, addi- tional antibody-mediated deprivation does not affect the speed of axonal regeneration of NGF-sensitive neurons [28] and certainly, although speculative, this could be due to a functionally stable state of NGF deprivation during the regeneration program, which coincides with a significant reduction in high affinity NGF receptors [29,30]. Intracellular signalling in the injured neuron In the injured neurons, the prompt arrival of signals for cellular injury and stress is followed by the induction of transcription factors, adhesion molecules, growth- associated proteins and structural components required for axonal elongation. Accompanying this is the activa- tion of intracellular signalling molecules, particularly molecules that control cell cycle and differentiation, synthesis of axonal transport molecules and cytoskele- ton components, secreted growth factors and cytokines, and a general increase in energy, amino acid and lipid M. Makwana and G. Raivich Molecular mechanism in regeneration FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 2629 metabolism [31–36]. Moreover, the introduction of the gene chip array technology has led to a dramatic increase in the number of identified genes regulated in injured and regenerating neurons [37–40] as well as the surrounding glia [41,42]. One of the earliest biochemical changes following axonal injury is the transient up-regulation of poly- amine-producing enzymes (ornithine decarboxylase, transglutaminase). This has been linked to an increase in mRNA metabolism and protein synthesis [43,44] resulting in polyamines such as putrescine, spermine and spermidine show a strong enhancing effect on neurite outgrowth both in vitro and in vivo [45–47]. Mitogen-activated protein (MAP) and phosphatidyl-inositol-3 kinase cascades Inhibition of ras, raf, the mitogen-associated protein kinase kinase (MEK) and the group of extracellular signal-regulated kinases 1 and 2 (erk1, erk2), molecules that are up-regulated after axonal injury [48], have been shown to inhibit the survival and neurite out- growth of embryonic neurons [49,50]. Conversely, studies in adult neurons suggest a loss or reversal of this pro-survival ⁄ pro-neurite outgrowth role [51–53], stressing the dissimilarity in the embryonic ⁄ neonatal and adult signalling. Increased activity of the ras ⁄ raf ⁄ mek pathway is complemented by a robust induction of phosphatidyl-inositol-3 kinase (PI3K) gene expres- sion [54], and the subsequent activation of protein kinase B or Akt [55,56]. Expression of active Akt also promotes axonal regeneration, verified using retrograde transport from peripheral target [57]; this effect may be associated with enhanced Akt-mediated distal branching observed in vitro [58]. Active Akt also enhances postnatal but not adult motoneuron survival; these effects are reversed by the expression of the dominant negative form of Akt [57]. Interestingly, the neuronal expression of constitutively activated V12-substituted ras enhances adult motoneuron survi- val, and does not result in the activation of Akt, but rather that of ERK1 and ERK2, the downstream targets of the ras ⁄ raf ⁄ mek pathway [59]. Transcription factors Following activation of MAP kinase pathways, phosphorylation and nuclear localization of a host of transcription factors including c-jun, junD, ATF3, P311, Sox11 and STAT3, as well as decreased expres- sion and activity of islet-1, ATF2 and nuclear factor kappa B [36,60–66], contribute to the change in gene expression of the injured neuron. In many cases, complete deletions for these transcrip- tion factors are embryonically lethal, limiting studies in vivo, in the adult animal. Nevertheless, recent advan- ces in cre ⁄ loxP technology has permitted cell type speci- fic, neuronal deletion of the loxP site flanked or floxed genes have begun to provide insight into the specific roles of these transcription factors. Neuronal specific deletion of STAT3 increases neuronal cell death after injury [21], to an degree similar as that observed for CNTF and LIF [67], thus pointing to the role of STAT3 as an intracellular survival-promoting factor. The effects of transcription factors on nerve regener- ation are well characterized in the case of c-jun. A recent study using the facial axotomy model showed that neur- onal deletion of c-jun hinders the expression of genes and proteins associated with axonal regeneration, redu- ces the speed of target reinnervation and functional recovery significantly and also completely blocks central axonal sprouting [68]. This is in keeping with previously demonstrated data [69–72] where deletion of jun blocks post-traumatic neuronal cell death and leads to severe neuronal atrophy. It also inhibits the recruitment of lymphocytes, and the activation of neighbouring micro- glia. Some axonal regeneration continues in the absence of c-jun highlighting the importance of alternative path- ways and compensatory mechanisms, which may be shared during developmental axonal outgrowth. These compensatory molecules may incorporate additional transcription factors such as STAT3 and ATF3 [73,74]; clarification of their role is needed to understand the signals regulating the gene expression switches occurring in the regenerating neuron. Cell death signals A number of studies point to presence of several cell surface molecules that mediate neuronal cell death following neonatal and adult axonal injury, including fas and tumour necrosis factor receptors (TNFR) 1 and 2 [75–77]. With the exception of TNFR2, these cell surface receptors carry a cytoplasmic death domain that exerts a pro-apoptotic signal through FADD. Furthermore, expression of dominant negative form of FADD has been shown to block the axotomy- induced cell death signal due to fas and TNFR [77]. Associated, downstream cytoplasmic cell death sig- nals play a key role in promoting neuronal cell death in neonatal and immature animals that are sensitive to axonal injury. Deletion of bax or inhibition of caspase 3 or the whole family of caspases has been shown to pre- vent cell death [78,79], but with an enhanced and more persistent effect for broad caspase inhibitors than caspase 3 alone [79]. In both cases, bax and caspases Molecular mechanism in regeneration M. Makwana and G. Raivich 2630 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS appear to act downstream of jun phosphorylation (jun-P) and the decrease in neuronal metabolism, suggesting a sequence of events beginning with jun-P, leading to atrophy, activation of bax and ending with the initiation of the caspase cascade. Unlike neonates, the effects of caspases in adult neurons are still poorly understood. For example, the global deletion of caspase 1, the interleukin (IL) 1 beta-converting enzyme, increa- ses the extent of motoneuron cell death by 40%, com- pared with wild-type controls [80]. Overall, the effects studied so far seem to be centred around neuronal survival, and the number of axons proximal to the injury site, although there was some indirect evidence for an enhancing effect of bax deletion on regeneration [78]. Inflammatory changes Axonal injury causes acute inflammatory changes at the lesion site, in the distal part of the nerve and around the cell body of injured neurons. In peripheral nerves, these changes are characterized by an influx of leukocytes that assist Schwann cells in clearing dys- functional myelin debris, around the cell body. These local inflammatory cells are rapidly activated and move into direct contact with the cell body to interact with T cells that patrol injured neural tissue [81]. In spite of these effects there appears to be little or moderate obvious correlation between the inflammatory changes described here and the speed of axonal regener- ation. Hence, deficiency for the macrophage colony- stimulating factor (MCSF) causes a severe reduction in microglial proliferation as well as early lymphocyte recruitment but does not affect the speed of axonal regeneration [82,83]. Similarly, a lack of effect on regen- eration has also been observed in mutant mice with enhanced inflammatory response to axonal injury, fol- lowing deletion of the common neurotrophin receptor p75NTR [84] or the protein tyrosine phosphatase shp1 [85]. Nevertheless, some studies do show a mild positive correlation. Gene deletion of IL 6 reduces inflammatory changes significantly around axotomized motoneurons [33], but also causes a moderate decrease in the speed of axonal regeneration [76,77]. Neurotrophic factors Numerous studies on injured peripheral neurons and neurotrophins have centred on the effects on post- traumatic neuronal survival, particularly in neonatal animals, where cell death is very pronounced [88–91]. In most cases, these studies showed a protective effect [20,92–95]. However, there were cases where externally applied neurotrophins reduced survival, particularly in the case of NGF; the toxic effect depended on the presence of the common neurotrophin receptor, the p75NTR [96]. A similar, toxic effect of p75NTR was observed on axonal regeneration in the sciatic [97] but not in the facial motor nerve [84]. Application of various neuro- trophins, growth factors and cytokines has also been demonstrated to encourage axonal outgrowth across the space between the disconnected, proximal and distal part of the axotomized peripheral nerve (reviewed in [98–101]). Studies concentrating on the speed of axonal regener- ation in the distal part of the nerve have thus far focussed on three groups of factors – the trophic factors insulin-like growth factor (IGF)-1, IGF2 and brain derived neurotrophic factor (BDNF), the transforming growth factor (TGF) beta superfamily member glial-cell derived neurotrophic factor (GDNF), and the neurokines leukaemia inhibitory factor (LIF) and IL 6. All three groups are implicated in the endo- genous regulation of the repair process. Application of the closely related exogenous growth factors IGF-1 and IGF-2 enhanced and application of antibodies reduced the pinch test-determined speed of axonal regeneration [102,103]. The combined overexpression of the neuro- kine IL 6 and its receptor resulted in improved nerve regeneration [104], whereas, genetic deletion of IL 6 has been shown to lead to a 15% reduction in the morphometrically determined speed of axonal regener- ation in the crushed facial motor nerve [87]. Similarly, a moderate effect was also observed using a functional, walking test assessment following sciatic nerve crush [105]. In contrast to this consistent but mild IL 6-medi- ated action in the periphery, there is substantial contro- versy regarding its effects on the CNS. Thus, deletion of IL6 has been shown to reduce glial scar formation and improve central axonal sprouting in the facial motor nucleus model [87], enhance functional recovery after spinal cord injury [106], but eliminate the condi- tional injury-induced spinal axon regeneration [107]. While both BDNF and GDNF are robustly and reliably induced in the distal portion of the injured peripheral nerve, the expression of their receptors on axotomized neurons slowly decreases during chronic disconnection [108]. Application of BDNF or GDNF did not improve regeneration immediately following injury, however, it promoted axonal regeneration of motoneurons whose regenerative ability has decreased by chronic axotomy 2 months prior to nerve resuture [97,109]. These delayed activities totally reversed the harmful effects of late nerve repair, which could indicate insufficient BDNF ⁄ GDNF-mediated support following chronic axotomy. These findings are in keeping with M. Makwana and G. Raivich Molecular mechanism in regeneration FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 2631 the strong, 50% reduction in the enduring success of the reinnervation of peripheral muscle in trkB + mice, where gene dosage of the primary BDNF receptor is also reduced by 50% [110]. Inhibition of endogenous BDNF [111] and GDNF [78] also decreases the postaxo- tomy collateral sprouting in the peripheral nerve. Cell adhesion molecules Functional studies in vivo have thus far centred on the role of laminin, its alpha7 beta1 integrin receptor and galectin. Deletion of the gene encoding alpha7 integrin subunit results in a severe, approximately 40% reduc- tion in the speed of motor axon regeneration following facial axotomy and a strong delay in the reinnervation of its peripheral target [112]. Neuronal jun-deficient mice do not show an up-regulation of alpha7 after injury, which appears to contribute to the very poor regenerative response seen in these animals [68]. The absence of laminin, the primary ligand for the alpha7 beta1 integrin, led to similar but more marked and lasting effects. Thus, cell type specific deletion of the gene for the gamma 1 laminin chain, an essential com- ponent of the laminin alpha beta gamma trimer in peripheral Schwann cells, caused a large decrease in the number of axons crossing into the distal portion of the crushed sciatic nerve for up to 30 days [113], in con- trast to the more transient defect in alpha7-deficient animals [112]. Interestingly, these alpha7-deficient mice show immense up-regulation of the beta1 integrin fol- lowing injury, which corresponds with a significant increase in central axonal sprouting [114] and may imply compensatory mechanisms involving other, clo- sely related alpha subunits. Central axonal sprouting on injury-induced neuronal cell surface molecules such as L1 and the close homologue of L1 (CHL1) relies on beta 1-integrin function [115–117], and may be involved in the effects observed in alpha7-deficient mice. In keeping with their trimeric and multimodal struc- ture, the various laminins can act as ligands for a plethora of different receptors, including the integrins, gicerin ⁄ CD146, the 67 kDa laminin-receptor, alpha- dystroglycan, and b-1,4-galactosyltransferase [118– 120]. Application of exogenous galectin-1 has been shown to promote the speed of neurite outgrowth, whereas the removal of endogenous galectin with anti- body neutralization or through gene deletion decreases it [121–123]. Galectin-1, in its oxidized form, also sti- mulates the migration of Schwann cells from the proxi- mal and distal stumps, assisting in the formation of cellular bridges permitting axonal growth into the distal part of the injured nerve. Cell surface interactions Normally, successful axonal regeneration is accompan- ied by the appearance of numerous, functionally diverse families of molecules that regulate surface–cytoskelet- elal interaction. The GAP43, MARCKS and CAP23 cytoplasmic proteins summarised by the acronym GMC [124–127], codistribute with the phosphoinositol- 4,5-diphosphate, PI(4,5)P2, at the semicrystalline, raft regions of the cell membrane [128]. These GMC molecules are functionally exchangeable, they bind exclusively to acidic phospholipids such as PI-(4,5)-P2, calcium ⁄ calmodulin, protein kinase C and actin fila- ments, and modify the raft-recruitment of signalling molecules such as src. They also regulate actin cyto- skeleton polymerization, organization and disassembly [129,130], and have a formative role in the appearance of filopodia and microspikes, in addition to the process of neurite outgrowth [127]. Interactions of calcium containing and noncontain- ing forms of calmodulin with the GMC molecules [131,132] offer an important connection for translating receptor-mediated calcium fluxes into signals guiding growth cone activity [133,134]. Inhibition of calcium influx in vivo with nimodipine clearly improves axonal regeneration [135,136]. Microtubule disassembly molecules such as SG10, stathmin and RB3, and associated counterparts – the collapsin response-mediated proteins, e.g. CRMP2 ) form a second, microtubule-targeting group of axonal adaptor molecules up-regulated after nerve injury [137–140]. Neuronal overexpression of CRMP2 has been shown to improve axonal regeneration [140]. These functions are supplemented by the microtubule associated protein 1b (MAP1B) that mediates the directionality of growth cone migration and axonal branching in the regeneration of sensory neurons [141]. The Rho GTPases, whose major members are RhoA, Rac, Cdc42 and TC10, form a third family of proteins that act as molecular switches which regulate cytoskele- tal structure, dynamics, and cell adhesion [142]. Acti- vation of these GTPases is mediated by adaptor molecules connected with cell surface receptors such as the slit-robo GTPase-activating protein 2 [143]. Axonal injury results in a small up-regulation of rhoA, rac, cdc42, and a massive increase in TC10 [144]. Inhibition of rhoA in vivo triggers a striking induction of the nor- mally absent axonal regeneration inside the CNS [1,145]. Overexpression of the cyclin-dependent-kinase inhibitor p21 (Cip1 ⁄ WAF1), which develops a complex with rho-kinase and inhibits its activity, also encour- ages regeneration and functional recovery in lesioned CNS [146]. Furthermore, molecules that stimulate the Molecular mechanism in regeneration M. Makwana and G. Raivich 2632 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS expression of p21, such as the nuclear localized protein p311, also promote regeneration in injured motor nerves, resulting in accelerated reinnervation of peri- pheral targets [36]. Conversely, not every cyclin- dependent kinase inhibits regeneration. Certainly, pharmacological blockade of cyclin-dependent kinase 5 with roscovitine or oloumucine has demonstrated a reduction in the ability of regenerating axons to over- come the moderate barrier to axonal elongation presen- ted by the lesion site of the crushed facial nerve [147]. Conclusion The aim of this review was to provide a synopsis on the various signals that are involved in mounting a successful regenerative response in the injured peri- pheral nervous system. It remains a complex process that involves a number of diverse and overlapping sig- nals. Particular emphasis was placed upon molecular signals and how this enables us to decipher the role of the endogenous molecules involved; in particular, recent data on specific neuronal gene deletions and antibody neutralization for specific proteins. Never- theless, continuing research permits us to overcome these hurdles and we are thus beginning to understand the complex processes involved in aiding us to repair the injured peripheral and central nervous system. References 1 Ellezam B, Dubreuil C, Winton M, Loy L, Dergham P, Selles-Navarro I & McKerracher L (2002) Inactiva- tion of intracellular Rho to stimulate axon growth and regeneration. Prog Brain Res 137, 371–380. 2 Fawcett J (2002) Repair of spinal cord injuries: where are we, where are we going? Spinal Cord 40, 615–623. 3 Schwab ME (2002) Increasing plasticity and functional recovery of the lesioned spinal cord. Prog Brain Res 137, 351–359. 4 Filbin MT (2003) Myelin-associated inhibitors of axo- nal regeneration in the adult mammalian CNS. Nat Rev Neurosci 4, 703–713. 5 Pekny M & Pekna M (2004) Astrocyte intermediate filaments in CNS pathologies and regeneration. J Pathol 204, 428–437. 6 Raivich G, Hellweg R & Kreutzberg GW (1991) NGF receptor-mediated reduction in axonal NGF uptake and retrograde transport following sciatic nerve injury and during regeneration. Neuron 7, 151–164. 7 Shadiack AM, Sun Y & Zigmond RE (2001) Nerve growth factor antiserum induces axotomy-like changes in neuropeptide expression in intact sympathetic and sensory neurons. J Neurosci 21, 363–371. 8 Sendtner M, Gotz R, Holtmann B & Thoenen H (1997) Endogenous ciliary neurotrophic factor is a lesion factor for axotomized motoneurons in adult mice. J Neurosci 17, 6999–7006. 9 Kirsch M, Terheggen U & Hofmann HD (2003) Ciliary neurotrophic factor is an early lesion-induced retro- grade signal for axotomized facial motoneurons. Mol Cell Neurosci 24, 130–138. 10 Lindholm D, Heumann R, Meyer M & Thoenen H (1987) Interleukin-1 regulates synthesis of nerve growth factor in non-neuronal cells of rat sciatic nerve. Nature 330, 658–659. 11 Yoo S, Nguyen MP, Fukuda M, Bittner GD & Fish- man HM (2003) Plasmalemmal sealing of transected mammalian neurites is a gradual process mediated by Ca(2+)-regulated proteins. J Neurosci Res 74, 541–551. 12 Berdan RC, Easaw JC & Wang R (1993) Alterations in membrane potential after axotomy at different dis- tances from the soma of an identified neuron and the effect of depolarisation on neurite outgrowth and calcium channel expression. J Neurophysiol 69, 151–164. 13 Spira ME, Oren R, Dormann A, Ilouz N & Lev S (2001) Calcium, protease activation, and cytoskeleton remodeling underlie growth cone formation and neuro- nal regeneration. Cell Mol Neurobiol 21, 591–604. 14 Zheng JQ, Kelly TK, Chang B, Ryazantsev S, Rajasek- aran AK, Martin KC & Twiss JL (2001) A functional role for intra-axonal protein synthesis during axonal regeneration from adult sensory neurons. J Neurosci 21, 9291–9303. 15 Ambron RT & Walters ET (1996) Priming events and retrograde injury signals: a new perspective on the cel- lular and molecular biology of nerve regeneration. Mol Neurobiol 13, 61–79. 16 Ambron RT, Dulin MF, Zhang XP, Schmied R & Walters ET (1995) Axoplasm enriched in a protein mobilized by nerve injury induces memory-like altera- tions in Aplysia neurons. J Neurosci 15, 3440–3446. 17 Hanz S, Perlson E, Willis D, Zheng JQ, Massarwa R, Huerta JJ, Koltzenburg M, Kohler M, van-Minnen J, Twiss JL & Fainzilber M (2003) Axoplasmic importins enable retrograde injury signaling in lesioned nerve. Neuron 40, 1095–1104. 18 Eckenstein FP, Shipley GD & Nishi R (1991) Acidic and basic fibroblast growth factors in the nervous sys- tem: distribution and differential alteration of levels after injury of central versus peripheral nerve. J Neu- rosci 11, 412–419. 19 Funakoshi H, Frisen J, Barbany G, Timmusk T, Zachrisson O, Verge VM & Persson H (1993) Differen- tial expression of mRNAs for neurotrophins and their receptors after axotomy of the sciatic nerve. J Cell Biol 123, 455–465. M. Makwana and G. Raivich Molecular mechanism in regeneration FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 2633 20 Sendtner M, Dittrich F, Hughes RA & Thoenen H (1994) Actions of CNTF and neurotrophins on degen- erating motoneurons: preclinical studies and clinical implications. J Neurol Sci 124 (Suppl.), 77–83. 21 Schweizer U, Gunnersen J, Karch C, Wiese S, Holtmann B, Takeda K, Akira S & Sendtner M (2002) Conditional gene ablation of Stat3 reveals differential signaling requirements for survival of motoneurons during development and after nerve injury in the adult. J Cell Biol 156, 287–297. 22 Rende M, Muir D, Ruoslahti E, Hagg T, Varon S & Manthorpe M (1992) Immunolocalization of ciliary neuronotrophic factor in adult rat sciatic nerve. Glia 5, 25–32. 23 Dobrea GM, Unnerstall JR & Rao MS (1992) The expression of CNTF message and immunoreactivity in the central and peripheral nervous system of the rat. Dev Brain Res 66, 209–219. 24 Perry VH & Brown MC (1992) Macrophages and nerve regeneration. Curr Opin Neurobiol 2, 679–682. 25 Kreutzberg and Raivich (2001) Inflammatory Responses Following Nerve Injury. Marcel Dekker, Inc., New York, Basel. 26 Ransohoff RM (2002) Universes in Delicate Balance: Chemokines and the Nervous System. Elsevier, New York, Amsterdam. 27 Gold BG, Storm-Dickerson T & Austin DR (1993) Regulation of the transcription factor c-JUN by nerve growth factor in adult sensory neurons. Neurosci Lett 154, 129–133. 28 Diamond J, Coughlin M, Macintyre L, Holmes M & Visheau B (1987) Evidence that endogenous beta nerve growth factor is responsible for the collateral sprouting, but not the regeneration, of nociceptive axons in adult rats. Proc Natl Acad Sci USA 84, 6596–6600. 29 Raivich G & Kreutzberg GW (1987) Expression of growth factor receptors in injured nervous tissue. I. Axotomy leads to a shift in the cellular distribution of specific beta-nerve growth factor binding in the injured and regenerating PNS. J Neurocytol 16, 689–700. 30 Verge VM, Riopelle RJ & Richardson PM (1989) Nerve growth factor receptors on normal and injured sensory neurons. J Neurosci 9, 914–922. 31 Livesey FJ, O’Brien JA, Li M, Smith AG, Murphy LJ & Hunt SP (1997) A Schwann cell mitogen accompanying regeneration of motor neurons. Nature 390, 614–618. 32 Su QN, Namikawa K, Toki H & Kiyama H (1997) Differential display reveals transcriptional up-regula- tion of the motor molecules for both anterograde and retrograde axonal transport during nerve regeneration. Eur J Neurosci 9, 1542–1547. 33 Klein MA, Moller JC, Jones LL, Bluethmann H, Kreutzberg GW & Raivich G (1997) Impaired neuro- glial activation in interleukin-6 deficient mice. Glia 19, 227–233. 34 Schmitt AB, Breuer S, Liman J, Buss A, Schlangen C, Pech K, Hol EM, Brook GA, Noth J & Schwaiger FW (2003) Identification of regeneration-associated genes after central and peripheral nerve injury in the adult rat. BMC Neurosci 4,8. 35 Vogelaar CF, Hoekman MF, Gispen WH & Burbach JP (2003) Homeobox gene expression in adult dorsal root ganglia during sciatic nerve regeneration: is regen- eration a recapitulation of development? Eur J Pharma- col 480, 233–250. 36 Fujitani M, Yamagishi S, Che YH, Hata K, Kubo T, Ino H, Tohyama M & Yamashita T (2004) P311 accel- erates nerve regeneration of the axotomized facial nerve. J Neurochem 91, 737–744. 37 Bonilla IE, Tanabe K & Strittmatter SM (2002) Small praline-rich repeat protein 1A is expressed by axotomised neurons and promotes axonal outgrowth. J Neurosci 22, 1303–1315. 38 Kubo T, Yamashita T, Yamaguchi A, Hosokawa K & Tohyama M (2002) Analysis of genes induced in peri- pheral nerve after axotomy using cDNA microarrays. J Neurochem 82, 1129–1136. 39 Boeshore KL, Schreiber RC, Vaccariello SA, Sachs HH, Salazar R, Lee J, Ratan RR, Leahy P & Zigmond RE (2004) Novel changes in gene expression following axotomy of a sympathetic ganglion: a microarray ana- lysis. J Neurobiol 59, 216–235. 40 Kury P, Abankwa D, Kruse F, Greine-Petter R & Muller HW (2004) Gene expression profiling reveals multiple novel intrinsic and extrinsic factors associated with axonal regeneration failure. Eur J Neurosci 19, 32–42. 41 Cameron AA, Vansant G, Wu W, Carlo DJ & Ill CR (2003) Identification of reciprocally regulated gene modules in regenerating dorsal root ganglion neurons and activated peripheral or central nervous system glia. J Cell Biochem 88, 970–985. 42 Moran LB, Duke DC, Turkheimer FE, Banati RB & Graeber MB (2004) Towards a transcriptome definition of microglial cells. Neurogenetics 5, 95–108. 43 Schwartz M, Kohsaka S & Agranoff BW (1981) Ornithine decarboxylase activity in retinal explants of goldfish undergoing optic nerve regeneration. Brain Res 227, 403–413. 44 Tetzlaff W, Gilad VH, Leonard C, Bisby MA & Gilad GM (1988) Retrograde changes in transglutaminase activity after peripheral nerve injuries. Brain Res 445, 142–146. 45 Sebille A & Bondoux-Jahan M (1980) Motor func- tion recovery after axotomy: enhancement by cyclo- phosphamide and spermine in rat. Exp Neurol 70, 507–515. 46 Schreiber RC, Boeshore KL, Laube G, Veh RW & Zigmond RE (2004) Polyamines increase in sympathetic neurons and non-neuronal cells after axotomy and Molecular mechanism in regeneration M. Makwana and G. Raivich 2634 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS enhance neurite outgrowth in nerve growth factor- primed PC12 cells. Neuroscience 128, 741–749. 47 Oble DA, Burton L, Maxwell K, Hassard T & Natha- niel EJ (2004) A comparison of thyroxine- and poly- amine-mediated enhancement of rat facial nerve regeneration. Exp Neurol 189, 105–111. 48 Kiryu S, Morita N, Ohno K, Maeno H & Kiyama H (1995) Regulation of mRNA expression involved in Ras and PKA signal pathways during rat hypoglossal nerve regeneration. Mol Brain Res 29, 147–156. 49 Borasio GD, John J, Wittinghofer A, Barde YA, Sendtner M & Heumann R (1989) ras p21 protein pro- motes survival and fiber outgrowth of cultured embryo- nic neurons. Neuron 2, 1087–1096. 50 Ihara S, Nakajima K, Fukada T, Hibi M, Nagata S, Hirano T & Fukui Y (1997) Dual control of neurite outgrowth by STAT3 and MAP kinase in PC12 cells stimulated with interleukin-6. EMBO J 16, 5345– 5352. 51 Liu RY & Snider WD (2001) Different signaling path- ways mediate regenerative versus developmental sen- sory axon growth. J Neurosci 21, RC164. 52 Wiklund P, Ekstrom PA & Edstrom A (2002) Mitogen- activated protein kinase inhibition reveals differences in signalling pathways activated by neurotrophin-3 and other growth-stimulating conditions of adult mouse dorsal root ganglia neurons. J Neurosci Res 67, 62–68. 53 Raivich G, Zirrgiebel U, Makwana M, Gschwendtner A, Kalla R, Miller FM & Kaplan D (2004) Mek pro- motes neuronal cell death and inhibits central sprouting following nerve injury. Soc Neurosci Abstr 390.5. 54 Ito Y, Sakagami H & Kondo H (1996) Enhanced gene expression for phosphatidylinositol 3-kinase in the hypoglossal motoneurons following axonal crush. Mol Brain Res 37, 329–332. 55 Owada Y, Utsunomiya A, Yoshimoto T & Kondo H (1997) Expression of mRNA for Akt, serine-threonine protein kinase, in the brain during development and its transient enhancement following axotomy of hypoglos- sal nerve. J Mol Neurosci 9, 27–33. 56 Murashov AKUI, Haq I, Hill C, Park E, Smith M, Wang X, Wang X, Goldberg DJ & Wolgemuth DJ (2001) Crosstalk between p38, Hsp25 and Akt in spinal motor neurons after sciatic nerve injury. Mol Brain Res 93, 199–208. 57 Namikawa K, Honma M, Abe K, Takeda M, Mansur K, Obata T, Miwa A, Okado H & Kiyama H (2000) Akt ⁄ protein kinase B prevents injury-induced motoneuron death and accelerates axonal regeneration. J Neurosci 20, 2875–2886. 58 Markus A, Zhong J & Snider WD (2002) Raf and Akt mediate distinct aspects of sensory axon growth. Neu- ron 35, 65–76. 59 Heumann R, Goemans C, Bartsch D, Lingenhohl K, Waldmeier PC, Hengerer B, Allegrini PR, Schellander K, Wagner EF, Arendt T, Kamdem RH, Obst-Pern- berg K, Narz F, Wahle P & Berns H (2000) Transgenic activation of Ras in neurons promotes hypertrophy and protects from lesion-induced degeneration. J Cell Biol 151, 1537–1548. 60 Herdegen T, Kummer W, Fiallos CE, Leah J & Bravo R (1991) Expression of c-JUN, JUN B and JUN D proteins in rat nervous system following transection of vagus nerve and cervical sympathetic trunk. Neurosci 45, 413–422. 61 Jenkins R & Hunt SP (1991) Long-term increase in the levels of c-jun mRNA and jun protein-like immuno- reactivity in motor and sensory neurons following axon damage. Neurosci Lett 129, 107–110. 62 Schwaiger FW, Hager G, Schmitt AB, Horvat A, Hager G, Streif R, Spitzer C, Gamal S, Breuer S, Brook GA, Nacimiento W & Kreutzberg GW (2000) Peripheral but not central axotomy induces changes in Janus kinases (JAK) and signal transducers and activa- tors of transcription (STAT). Eur J Neurosci 12, 1165– 1176. 63 Mason MR, Lieberman AR & Anderson PN (2003) Corticospinal neurons up-regulate a range of growth- associated genes following intracortical, but not spinal, axotomy. Eur J Neurosci 18, 789–802. 64 Tanabe K, Bonilla I, Winkles JA & Strittmatter SM (2003) Fibroblast growth factor-inducible-14 is induced in axotomized neurons and promotes neurite out- growth. J Neurosci 23, 9675–9686. 65 Doyle CA & Hunt SP (1997) Reduced nuclear factor kappaB (p65) expression in rat primary sensory neurons after peripheral nerve injury. Neuroreport 8, 2937–2942. 66 Povelones M, Tran K, Thanos D & Ambron RT (1997) An NF-kappaB-like transcription factor in axo- plasm is rapidly inactivated after nerve injury in Aply- sia. J Neurosci 17, 4915–4920. 67 Sendtner M, Gotz R, Holtmann B, Escary JL, Masu Y, Carroll P, Wolf E, Brem G, Brulet P & Thoenen H (1996) Cryptic physiological trophic support of moto- neurons by LIF revealed by double gene targeting of CNTF and LIF. Curr Biol 6, 686–694. 68 Raivich G, Bohatschek M, Da Costa C, Iwata O, Galiano M, Hristova M, Nateri AS, Makwana M, Riera-Sans L, Wolfer DP, Lipp HP, Aguzzi A, Wagner EF & Behrens A (2004) The AP-1 transcription factor c-Jun is required for efficient axonal regeneration. Neuron 43, 57–67. 69 Ham J, Babij C, Whitfield J, Pfarr CM, Lallemand D, Yaniv M & Rubin LL (1995) A c-Jun dominant nega- tive mutant protects sympathetic neurons against pro- grammed cell death. Neuron 14, 927–939. 70 Xia Z, Dickens M, Raingeaud J, Davis RJ & Greenberg ME (1995) Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 270, 1326–1331. M. Makwana and G. Raivich Molecular mechanism in regeneration FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 2635 71 Palmada M, Kanwal S, Rutkoski NJ, Gustafson- Brown C, Johnson RS, Wisdom R & Carter BD (2002) c-jun is essential for sympathetic neuronal death induced by NGF withdrawal but not by p75 activation. J Cell Biol 158, 453–461. 72 Lindwall C, Dahlin L, Lundborg G & Kanje M (2004) Inhibition of c-Jun phosphorylation reduces axonal outgrowth of adult rat nodose ganglia and dorsal root ganglia sensory neurons. Mol Cell Neurosci 27, 267– 279. 73 Pearson AG, Gray CW, Pearson JF, Greenwood JM, During MJ & Dragunow M (2003) ATF3 enhances c-Jun – mediated neurite sprouting. Mol Brain Res 120, 38–45. 74 Zhou FQ, Walzer MA & Snider WD (2004) Turning on the machine: genetic control of axon regeneration by c-Jun. Neuron 43, 1–2. 75 Terrado J, Monnier D, Perrelet D, Vesin D, Jemelin S, Buurman WA, Mattenberger L, King B, Kato AC & Garcia I (2000) Soluble TNF receptors partially protect injured motoneurons in the postnatal CNS. Eur J Neurosci 12, 3443–3447. 76 Raivich G, Liu ZQ, Kloss CU, Labow M, Bluethmann H & Bohatschek M (2002) Cytotoxic potential of proinflammatory cytokines: combined deletion of TNF receptors TNFR1 and TNFR2 prevents motoneuron cell death after facial axotomy in adult mouse. Exp Neurol 178, 186–193. 77 Ugolini G, Raoul C, Ferri A, Haenggeli C, Yamamoto Y, Salaun D, Henderson CE, Kato AC, Pettmann B & Hueber AO (2003) Fas ⁄ tumor necrosis factor receptor death signaling is required for axotomy-induced death of motoneurons in vivo. J Neurosci 23, 8526–8531. 78 Sun W & Oppenheim RW (2003) Response of moto- neurons to neonatal sciatic nerve axotomy in Bax- knockout mice. Mol Cell Neurosci 24, 875–886. 79 Chan YM, Yick LW, Yip HK, So KF, Oppenheim RW & Wu W (2003) Inhibition of caspases promotes long-term survival and reinnervation by axotomized spinal motoneurons of denervated muscle in newborn rats. Exp Neurol 181, 190–203. 80 de Bilbao F, Giannakopoulos P, Srinivasan A & Dubois-Dauphin M (2000) In vivo study of motoneuron death induced by nerve injury in mice deficient in the caspase 1 ⁄ interleukin-1 beta-converting enzyme. Neu- roscience 98, 573–583. 81 Kloss CU, Werner A, Klein MA, Shen J, Menuz K, Probst JC, Kreutzberg GW & Raivich G (1999) Integrin family of cell adhesion molecules in the injured brain: regulation and cellular localization in the normal and regenerating mouse facial motor nucleus. J Comp Neurol 411, 162–178. 82 Raivich G, Moreno-Flores MT, Moller JC & Kreutz- berg GW (1994) Inhibition of posttraumatic microglial proliferation in a genetic model of macrophage colony-stimulating factor deficiency in the mouse. Eur J Neurosci 6, 1615–1618. 83 Kalla R, Liu Z, Xu S, Koppius A, Imai Y, Kloss CU, Kohsaka S, Gschwendtner A, Moller JC, Werner A & Raivich G (2001) Microglia and the early phase of immune surveillance in the axotomized facial motor nucleus: impaired microglial activation and lymphocyte recruitment but no effect on neuronal survival or axo- nal regeneration in macrophage-colony stimulating fac- tor-deficient mice. J Comp Neurol 436, 182–201. 84 Gschwendtner A, Liu Z, Hucho T, Bohatschek M, Kalla R, Dechant G & Raivich G (2003) Regulation, cellular localization, and function of the p75 neurotro- phin receptor (p75NTR) during the regeneration of facial motoneurons. Mol Cell Neurosci 24, 307–322. 85 Horvat A, Schwaiger F, Hager G, Brocker F, Streif R, Knyazev P, Ullrich A & Kreutzberg GW (2001) A novel role for protein tyrosine phosphatase shp1 in controlling glial activation in the normal and injured nervous system. J Neurosci 21, 865–874. 86 Zhong J, Dietzel ID, Wahle P, Kopf M & Heumann R (1999) Sensory impairments and delayed regeneration of sensory axons in interleukin-6-deficient mice. J Neu- rosci 19 , 4305–4313. 87 Galiano M, Liu ZQ, Kalla R, Bohatschek M, Koppius A, Gschwendtner A, Xu S, Werner A, Kloss CU, Jones LL, Bluethmann H & Raivich G (2001) Interleukin-6 (IL6) and cellular response to facial nerve injury: effects on lymphocyte recruitment, early microglial activation and axonal outgrowth in IL6-deficient mice. Eur J Neu- rosci 14 , 327–341. 88 Miyata Y, Kashihara Y, Homma S & Kuno M (1986) Effects of nerve growth factor on the survival and synaptic function of Ia sensory neurons axotomized in neonatal rats. J Neurosci 6, 2012–2018. 89 Obouhova G, Clowry GJ & Vrbova G (1994) Changes in retrogradely labelled neurones in the red nucleus and cortex after depletion of motoneurones by axotomy in neonatal rats. Dev Neurosci 16, 34–37. 90 Moran LB & Graeber MB (2004) The facial nerve axotomy model. Brain Res Rev 44, 154–178. 91 Schmalbruch H (1987) Loss of sensory neurons after sciatic nerve section in the rat. Anat Rec 219, 323–329. 92 Sendtner M, Kreutzberg GW & Thoenen H (1990) Ciliary neurotrophic factor prevents the degeneration of motor neurons after axotomy. Nature 345, 440–441. 93 Yan Q, Elliott J & Snider WD (1992) Brain-derived neurotrophic factor rescues spinal motor neurons from axotomy-induced cell death. Nature 360, 753–755. 94 Koliatsos VE, Clatterbuck RE, Winslow JW, Cayou- ette MH & Price DL (1993) Evidence that brain- derived neurotrophic factor is a trophic factor for motor neurons in vivo. Neuron 10, 359–367. Molecular mechanism in regeneration M. Makwana and G. Raivich 2636 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 95 Arenas E & Persson H (1994) Neurotrophin-3 prevents the death of adult central noradrenergic neurons in vivo. Nature 367, 368–371. 96 Wiese S, Metzger F, Holtmann B & Sendtner M (1999) The role of p75NTR in modulating neurotrophin survi- val effects in developing motoneurons. Eur J Neurosci 11, 1668–1676. 97 Boyd JG & Gordon T (2002) A dose-dependent facili- tation and inhibition of peripheral nerve regeneration by brain-derived neurotrophic factor. Eur J Neurosci 15, 613–626. 98 Varon S, Manthorpe M & Williams LR (1983) Neuro- notrophic and neurite-promoting factors and their clini- cal potentials. Dev Neurosci 6, 73–100. 99 Raivich G & Kreutzberg GW (1993) Peripheral nerve regeneration: role of growth factors and their receptors. Int J Dev Neurosci 11, 311–324. 100 Terenghi G (1999) Peripheral nerve regeneration and neurotrophic factors. J Anat 194, 1–14. 101 Boyd JG & Gordon T (2003a) Neurotrophic factors and their receptors in axonal regeneration and func- tional recovery after peripheral nerve injury. Mol Neu- robiol 27, 277–324. 102 Kanje M, Skottner A, Sjoberg J & Lundborg G (1989) Insulin-like growth factor I (IGF-I) stimulates regen- eration of the rat sciatic nerve. Brain Res 486, 396–398. 103 Glazner GW, Lupien S, Miller JA & Ishii DN (1993) Insulin-like growth factor II increases the rate of sciatic nerve regeneration in rats. Neuroscience 54, 791–797. 104 Hirota H, Kiyama H, Kishimoto T & Taga T (1996) Accelerated nerve regeneration in mice by upregulated expression of interleukin 6 and IL-6 receptor after trauma. J Exp Med 183, 2627–2634. 105 Inserra MM, Yao M, Murray R & Terris DJ (2000) Peripheral nerve regeneration in interleukin 6-deficient mice. Arch Otolaryngol Head Neck Surg 126, 1112– 1116. 106 Okada S, Nakamura M, Mikami Y, Shimazaki T, Mihara M, Ohsugi Y, Iwamoto Y, Yoshizaki K, Kishimoto T, Toyama Y & Okano H (2004) Blockade of interleukin-6 receptor suppresses reactive astrogliosis and ameliorates functional recovery in experimental spinal cord injury. J Neurosci Res 76, 265–276. 107 Cafferty WB, Gardiner NJ, Das P, Qiu J, McMahon SB & Thompson SW (2004) Conditioning injury- induced spinal axon regeneration fails in interleukin-6 knock-out mice. J Neurosci 24, 4432–4443. 108 Hammarberg H, Piehl F, Risling M & Cullheim S (2000) Differential regulation of trophic factor receptor mRNAs in spinal motoneurons after sciatic nerve transection and ventral root avulsion in the rat. J Comp Neurol 426, 587–601. 109 Boyd JG & Gordon T (2003b) Glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor sustain the axonal regeneration of chronically axotomized motoneurons in vivo. Exp Neurol 183, 610– 619. 110 Boyd JG & Gordon T (2001) The neurotrophin recep- tors, trkB and p75, differentially regulate motor axonal regeneration. J Neurobiol 49, 314–325. 111 Streppel M, Azzolin N, Dohm S, Guntinas-Lichius O, Haas C, Grothe C, Wevers A, Neiss WF & Angelov DN (2002) Focal application of neutralizing antibodies to soluble neurotrophic factors reduces collateral axonal branching after peripheral nerve lesion. Eur J Neurosci 15, 1327–1342. 112 Werner A, Willem M, Jones LL, Kreutzberg GW, Mayer U & Raivich G (2000) Impaired axonal regen- eration in alpha7 integrin-deficient mice. J Neurosci 20, 1822–1830. 113 Chen ZL & Strickland S (2003) Laminin gamma1 is critical for Schwann cell differentiation, axon myelination, and regeneration in the peripheral nerve. J Cell Biol 163, 889–899. 114 Werner A, Mayer U, Kreutzberg GW & Raivich G (1998b) Molecular constituents of regenerating growth cones of mouse motoneurons. Clin Neuropathol 17, 285. 115 Buhusi M, Midkiff BR, Gates AM, Richter M, Schach- ner M & Maness PF (2003) Close homolog of L1 is an enhancer of integrin-mediated cell migration. J Biol Chem 278, 25024–25031. 116 Chaisuksunt V, Campbell G, Zhang Y, Schachner M, Lieberman AR & Anderson PN (2003) Expression of regeneration-related molecules in injured and regenerat- ing striatal and nigral neurons. J Neurocytol 32, 161– 183. 117 Becker CG, Lieberoth BC, Morellini F, Feldner J, Becker T & Schachner M (2004) L1.1 is involved in spinal cord regeneration in adult zebrafish. J Neurosci 24, 7837–7842. 118 Ekblom P, Lonai P & Talts JF (2003) Expression and biological role of laminin-1. Matrix Biol 22, 35–47. 119 Powell SK & Kleinman HK (1997) Neuronal laminins and their cellular receptors. Int J Biochem Cell Biol 29, 401–414. 120 Taira E, Takaha N, Taniura H, Kim CH & Miki N (1994) Molecular cloning and functional expression of gicerin, a novel cell adhesion molecule that binds to neurite outgrowth factor. Neuron 12, 861–872. 121 Horie H & Kadoya T (2000) Identification of oxidized galectin-1 as an initial repair regulatory factor after axotomy in peripheral nerves. Neurosci Res 38, 131– 137. 122 Fukaya K, Hasegawa M, Mashitani T, Kadoya T, Horie H, Hayashi Y, Fujisawa H, Tachibana O, Kida S & Yamashita J (2003) Oxidized galectin-1 stimulates the migration of Schwann cells from both proximal and distal stumps of transected nerves and M. Makwana and G. Raivich Molecular mechanism in regeneration FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS 2637 [...]... Yoneda Y, Ochi T, Tohyama M, Yoshikawa H & Kiyama H (2000) The small GTP-binding protein TC10 promotes nerve elongation in neuronal cells, and its expression is induced during nerve regeneration in rats Neurosci 20, 4138–4144 145 Fournier AE, Takizawa BT & Strittmatter SM (2003) Rho kinase inhibition enhances axonal regeneration in the injured CNS J Neurosci 23, 1416–1423 146 Tanaka H, Yamashita T, Yachi... growth-associated polypeptides in regenerating toad retinal ganglion cell axons J Neurosci 1, 419–426 Verhaagen J, van Hooff CO, Edwards PM, De Graan PN, Oestreicher AB, Schotman P, Jennekens FG & Gispen WH (1986) The kinase C substrate protein B-50 and axonal regeneration Brain Res Bull 17, 737–741 McNamara RK, Jiang Y, Streit WJ & Lenox RH (2000) Facial motor neuron regeneration induces a unique spatial... temporal pattern of myristoylated alanine-rich C kinase substrate expression Neuroscience 97, 581–589 Bomze HM, Bulsara KR, Iskandar BJ, Caroni P & Skene JH (2001) Spinal axon regeneration evoked by replacing two growth cone proteins in adult neurons Nat Neurosci 4, 38–43 Frey D, Laux T, Xu L, Schneider C & Caroni P (2000) Shared and unique roles of CAP23 and GAP43 in actin regulation, neurite outgrowth,... molecules during nerve regeneration Mol Brain Res 102, 105–109 138 Mori N & Morii H (2002) SCG10-related neuronal growth-associated proteins in neural development, plasticity, degeneration, and aging J Neurosci Res 70, 264–273 139 Mason MR, Lieberman AR, Grenningloh G & Anderson PN (2002) Transcriptional upregulation of SCG10 and CAP-23 is correlated with regeneration of the axons of peripheral and... cone migration and axonal branching in regeneration of adult dorsal root ganglia neurons J Neurosci 24, 7204– 7213 142 Etienne-Manneville S & Hall A (2002) Rho GTPases in cell biology Nature 420, 629–635 143 Madura T, Yamashita T, Kubo T, Tsuji L, Hosokawa K & Tohyama M (2004) Changes in mRNA of SlitRobo GTPase-activating protein 2 following facial nerve transection Mol Brain Res 123, 76–80 144 Tanabe.. .Molecular mechanism in regeneration 123 124 125 126 127 128 129 130 131 132 133 134 135 promotes axonal regeneration after peripheral nerve injury J Neuropathol Exp Neurol 62, 162–172 McGraw J, McPhail LT, Oschipok LW, Horie H, Poirier F, Steeves JD, Ramer MS & Tetzlaff W (2004) Galectin-1 in regenerating motoneurons Eur J Neurosci 20, 2872–2880 Skene... Fujiwara T, Yoshikawa H & Tohyama M (2004) Cytoplasmic p21 (Cip1 ⁄ WAF1) enhances axonal regeneration and functional recovery after spinal cord injury in rats Neuroscience 127, 155–164 147 Namgung U, Choi BH, Park S, Lee JU, Seo HS, Suh BC & Kim KT (2004) Activation of cyclin-dependent kinase 5 is involved in axonal regeneration Mol Cell Neurosci 25, 422–432 FEBS Journal 272 (2005) 2628–2638 ª 2005 FEBS... neurons in vivo Mol Cell Neurosci 20, 595–615 140 Suzuki Y, Nakagomi S, Namikawa K, Kiryu-Seo S, Inagaki N, Kaibuchi K, Aizawa H, Kikuchi K & Kiyama H (2003) Collapsin response mediator protein2 accelerates axon regeneration of nerve-injured motor neurons of rat J Neurochem 86, 1042–1050 141 Bouquet C, Soares S, von Boxberg Y, Ravaille-Veron M, Propst F & Nothias F (2004) Microtubule-associated protein... (1996) Nimodipine accelerates axonal sprouting after surgical repair of rat facial nerve J Neurosci 16, 1041–1048 2638 M Makwana and G Raivich 136 Mattsson P, Janson AM, Aldskogius H & Svensson M (2001) Nimodipine promotes regeneration and functional recovery after intracranial facial nerve crush J Comp Neurol 437, 106–117 137 Iwata T, Namikawa K, Honma M, Mori N, Yachiku S & Kiyama H (2002) Increased... PI(4,5)P(2) at plasmalemmal rafts, and regulate cell cortex actin dynamics through a common mechanism J Cell Biol 149, 1455–1472 Caroni P (2001) Actin cytoskeleton regulation through modulation of PI(4,5)P(2) rafts EMBO J 20, 4332– 4336 Gerendasy D (1999) Homeostatic tuning of Ca2+ signal transduction by members of the calpacitin protein family J Neurosci Res 58, 107–119 Dunican DJ & Doherty P (2000) . protein p311, also promote regeneration in injured motor nerves, resulting in accelerated reinnervation of peri- pheral targets [36]. Conversely, not every cyclin- dependent kinase inhibits regeneration. . GTPase-activating protein 2 [143]. Axonal injury results in a small up-regulation of rhoA, rac, cdc42, and a massive increase in TC10 [144]. Inhibition of rhoA in vivo triggers a striking induction. metabolism and protein synthesis [43,44] resulting in polyamines such as putrescine, spermine and spermidine show a strong enhancing effect on neurite outgrowth both in vitro and in vivo [45–47]. Mitogen-activated

Ngày đăng: 30/03/2014, 16:20

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan