Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Feb 1.
Published in final edited form as: Exp Neurol. 2007 May 31;209(2):294–301. doi: 10.1016/j.expneurol.2007.05.014

CNS Injury, Glial Scars, and Inflammation

Inhibitory extracellular matrices and regeneration failure

Michael T Fitch 1, Jerry Silver 2
PMCID: PMC2268907  NIHMSID: NIHMS40614  PMID: 17617407

Abstract

Spinal cord and brain injuries lead to complex cellular and molecular interactions within the central nervous system in an attempt to repair the initial tissue damage. Many studies have illustrated the importance of the glial cell response to injury, and the influences of inflammation and wound healing processes on the overall morbidity and permanent disability that result. The abortive attempts of neuronal regeneration after spinal cord injury are influenced by inflammatory cell activation, reactive astrogliosis and the production of both growth promoting and inhibitory extracellular molecules. Despite the historical perspective that the glial scar was a mechanical barrier to regeneration, inhibitory molecules in the forming scar and methods to overcome them have suggested molecular modification strategies to allow neuronal growth and functional regeneration. Unlike myelin associated inhibitory molecules, which remain at largely static levels before and after central nervous system trauma, inhibitory extracellular matrix molecules are dramatically upregulated during the inflammatory stages after injury providing a window of opportunity for the delivery of candidate therapeutic interventions. While high dose methylprednisolone steroid therapy alone has not proved to be the solution to this difficult clinical problem, other strategies for modulating inflammation and changing the make up of inhibitory molecules in the extracellular matrix are providing robust evidence that rehabilitation after spinal cord and brain injury has the potential to significantly change the outcome for what was once thought to be permanent disability.

Keywords: Astrocytes, glial scar, spinal injury, regeneration, inflammation, proteoglycan, extracellular matrix

Introduction

Meaningful rehabilitation after spinal cord injury encompasses many aspects of acute and chronic patient care. The underlying basic science of wound healing in the central nervous system (CNS) involves fundamental concepts of cellular structure and function, communication between glial cells and neuronal processes, and complex molecular interactions between cells and their extracellular environments. Understanding the basic functions of glial cells in the brain and spinal cord and their interactions with inflammatory cells and injured axons will allow the development of treatment strategies that can promote regeneration after injury (Fitch and Silver, 2007).

The glial cells of the CNS (astrocytes, oligodendrocytes and their precursors, and microglia) supply both structural and physiological support, and also respond to injury or disease. Oligodendrocytes provide myelin sheaths for enhanced axonal transmission (Wood and Bunge, 1984) but they or their progenitors can also remyelinate under certain circumstances (Chang et al., 2000; Nishiyama, 2007; Trotter, 2005; Wilson et al., 2006). Microglial cells are the resident immune system phagocytic cells within the brain and spinal cord (Kim and de Vellis, 2005), and they also respond rapidly for neural protection or healing after injury (Davalos et al., 2005; Wu et al., 2007b). Astrocytes are important for neurotransmitter regulation (Schousboe and Westergaarde, 1995), ion homeostasis (Walz, 1989), blood brain barrier maintenance (Wolburg and Risau, 1995), and the production of extracellular matrix molecules destined for the basal lamina and perineuronal net (Ard et al., 1993; Bernstein et al., 1985; Canning et al., 1996; Grierson et al., 1990; Massey et al., 2006; McKeon et al., 1991; Smith-Thomas et al., 1994; Tom et al., 2004a). After injury, they are also the major cell type responsible for walling off areas of damage to protect the fragile brain tissue from further erosion (Fitch et al., 1999; Myer et al., 2006). The formation of a mechanically obstructive glial scar composed of astrocytes and connective tissue elements was at one time thought to explain the failure of regeneration within the CNS after injury (Windle and Chambers, 1950). However, subsequent work has demonstrated that the glial scar is far more than just a physical barrier, as the role of glial cells in preventing successful regeneration involves complex cellular and molecular interactions (Fitch and Silver, 2001). Astrocytes, oligodendrocyte progenitor cells, microglia, and peripheral inflammatory cells with their proposed roles in the failure of functional regeneration in the CNS will be discussed in this article.

Nonpermissive Environment for Regeneration in the CNS

Growth Potential of CNS Neurons

Despite early claims that axons in the adult CNS were incapable of regeneration (Le Gros Clark, 1943), experimental research has demonstrated that CNS axons are capable of at least small amounts of regeneration. Sprouting within the lesion site (Liu and Chambers, 1958), (Guth et al., 1985), and even short distance sprouting from surviving axons (Freund et al., 2006; Goshgarian, 2003; Li and Raisman, 1994; Taylor et al., 2006) have been seen in several different experimental models. However, at the lesion site itself newly formed growth cones quickly become dystrophic (Li and Raisman, 1995; Misgeld and Kerschensteiner, 2006; Misgeld et al., 2007). They do not leave the immediate site of injury and appear incapable, when left to their own devices, of producing any long-distance regeneration.

Recent work has shown that a gradient of inhibitory extracellular matrix molecules can convert an actively extending axonal growth cone into a dystrophic ending that fails to continue long-distance regeneration. These sprouting tips of severed axons then remain stalled in place without further functional growth. Interestingly, these dystrophic growth cones are actually dynamic structures with continuing turnover of membrane components and are extremely dynamic in vitro and in vivo (Kerschensteiner et al., 2005; Tom et al., 2004b). Time-lapse video of these dystrophic endings illustrates repeated episodes of endocytosis and efforts to send out membrane veils (Tom, et al., 2004b). The failure of these dystrophic growth cones to continue growing to produce long-distance regeneration appears to be related to the inhibitory and nonpermissive nature of the injured brain and spinal cord.

In contrast to the CNS, the peripheral nervous system (PNS) is growth supporting for regeneration, especially after a crush injury (Lazar et al., 2004). Segments of peripheral nerves with the associated PNS environment can even encourage remarkable long-distance regeneration of many types of CNS axons when such grafts are inserted centrally (David and Aguayo, 1981; Richardson et al., 1980). These classic transplantation experiments proved that adult CNS axons do have an intrinsic capacity for lengthy regeneration, albeit perhaps less than optimal growth (see article by Filbin, et al., in this issue). However, the vast majority of regenerating axons fail to continue upon re-entering the CNS environment, which further demonstrates the inhibitory and nonpermissive nature of the CNS. The injury responses of the two branches of the sensory dorsal root ganglion (DRG) neuron further demonstrate the dichotomy between CNS and PNS regeneration, as successful long-distance regeneration easily occurs after injury to the peripheral branch but does not occur with attempted growth of the central branch into the CNS (Golding et al., 1997; Kliot et al., 1990).

Molecules Within the Glial Scar Contribute to Regenerative Failure

The formation of a histologically apparent glial scar, composed of astrocytes and connective tissue elements, is one of the most studied but poorly understood barriers to regeneration of CNS axons. Traditionally, thought to be a simple mechanical barrier (Windle and Chambers, 1950), later studies suggested that regeneration still fails even when a dense glial scar does not form (Guth et al., 1986). Multiple models have now demonstrated that the molecular composition of the scar and the production of inhibitory molecules by astrocytes are contributing factors for regenerative failure (Busch and Silver, 2007; Fawcett, 2006; Fitch and Silver, 1997a; Fitch and Silver, 2000; Liu et al., 2006; McGraw et al., 2001; Silver and Miller, 2004; Yiu and He, 2006; Zhang et al., 2006). Reactive astrocytes within the glial scar have been shown to upregulate molecules such as tenascin (Apostolova et al., 2006; Brodkey et al., 1995; McKeon et al., 1995), Semaphorin 3 (Pasterkamp et al., 2001), ephrin-B2 (Bundesen et al., 2003), slit proteins (Hagino et al., 2003), and a host of chondroitin sulfate proteoglycans (Jones et al., 2003a; McKeon, et al., 1995; Rhodes and Fawcett, 2004). While not all of these candidates have been fully characterized in terms of their contributions to regenerative failure, a substantial body of work has been accumulated to illustrate the inhibitory effects of scar formation within the CNS. Compelling evidence that the lesion environment itself is critical for CNS regenerative failure came from experiments where adult dorsal root ganglion neurons were micro-transplanted into undamaged or pre-lesioned white matter tracts (Davies et al., 1997; Davies et al., 1999). This careful micro-transplantation avoids a vigorous inflammatory response, which allows dorsal root ganglion neurons immediate access either to a relatively intact host glial environment or one that is undergoing Wallerian degeneration. These sensory axons regenerate rapidly and over long-distances within adult white matter tracts of the brain and spinal cord. As the rapidly growing adult neurons reach an area of CNS damage, with associated inflammatory infiltrates and inhibitory molecules, the axons convert into a dystrophic state and are unable to continue (Davies, et al., 1999). These findings highlight the importance of the glial scar in the creation of an inhibitory environment for unsuccessful regeneration within the CNS.

Increases in proteoglycan molecules have been found in the CNS in many different injury models. The NG2 proteoglycan is increased after CNS injury by oligodendrocyte precursor cells (Levine, 1994; Rhodes et al., 2006), phosphacan is upregulated within glial scars (McKeon, et al., 1995), and phosphacan, brevican, versican, and neurocan are all produced within the injured spinal cord (Jones, et al., 2003a). Chondroitin sulfate proteoglycans are found in the brain after stab wound (Fitch and Silver, 1997a), in the spinal cord after injury to the dorsal root (Pindzola et al., 1993), and in the spinal cord following injury (Fitch and Silver, 1997a; Jones, et al., 2003a; Jones et al., 2003b). The rapid and long lasting upregulation of proteoglycans within the vicinity of the glial scar has implicated them in the creation of nonpermissive growth environments in the CNS, similar to their role in boundary formation within the CNS during development (Fitch and Silver, 1997b; Grimpe and Silver, 2004; Silver, 1994; Snow et al., 1990). It is now well established that proteoglycans associated with reactive astrocytes clearly inhibit neurite outgrowth in vitro (Bovolenta et al., 1993; Canning et al., 1993; Dou and Levine, 1994; McKeon, et al., 1991; Snow et al., 1990; Tom, et al., 2004b), and these molecules also appear to play a key role in creating an environment that is not appropriate for successful long-distance regeneration of adult neurons after injury in vivo, since their modification allows successful regeneration to occur (Bradbury et al., 2002; Houle et al., 2006; Steinmetz et al., 2005).

Triggers for the Production of Inhibitory Extracellular Matrix

Unlike myelin-associated inhibitory molecules, which are present at relatively static levels before and after CNS injury (see Zheng, et al., in this issue), inhibitory matrices such as proteoglycans are upregulated in areas of gliosis following traumatic injuries in the brain and spinal cord. They are typically enhanced in regions of BBB breakdown (Fitch and Silver, 1997a; Rhodes, et al., 2006), but new evidence also suggests they are upregulated in the perineuronal net in areas distant from the lesion in synaptic centers denervated by the injury (Massey, et al., 2006). In order to appropriately manipulate the extracellular matrix, we must first consider those triggers that initiate the increased production of inhibitory molecules.

The cellular hypertrophy and increases in glial fibrillary acidic protein (GFAP) that are the hallmarks of astrocyte reactivity occur rapidly after CNS injury. While the specific triggers remain unclear, there are a number of hypotheses about factors that contribute to astrocyte activation and gliosis. Degeneration of severed axon tracts appears to lead to gliosis, even in regions remote from the site of trauma in the brain or spinal cord (Barrett et al., 1981; Fitch and Silver, 1997a; Massey, et al., 2006; Murray et al., 1990; Steward and Trimmer, 1997). Cytokines or other molecules that may trigger gliosis include TNF-alpha (Rostworowski et al., 1997), endothelin-1 (Hama et al., 1997), IL-1 (Giulian and Lachman, 1985), IL-6 (Chiang et al., 1994), thrombin (Nishino et al., 1993), and CNTF (Kahn et al., 1995). Some of these may originate as soluble serum factors, or they can be directly produced by astrocytes, activated microglia, or peripheral macrophages. Oligodendrocyte precursor cells also become activated in response to similar triggers such as platelets, macrophages, and inflammation-associated cytokines (Rhodes, et al., 2006).

Traditional definitions of astrocyte activation are based on morphology, numbers, and increases in GFAP expression and/or detection. However, it is perhaps more important to consider which triggers lead not only to “gliosis” but to the production of inhibitory molecules such as proteoglycans that create a nonpermissive environment for axons attempting to regenerate. Some have suggested that degenerating axons and their dying terminals may be a trigger for such extracellular matrix production by astrocytes (see Massey, et al., in this issue). Evidence also suggests that the inflammatory infiltration and activation associated with CNS injury is associated with production of inhibitory molecules, as CSPGs are associated with breakdown of the blood brain barrier and infiltrating macrophages present within a lesion site (Fitch, et al., 1999; Fitch and Silver, 1997a), a phenomenon that is also seen with activation of oligodendrocyte precursor cells after injury (Rhodes, et al., 2006). Pro-inflammatory molecules that activate microglial cells and macrophages can trigger astrocyte gliosis and upregulation of proteoglycans (Fitch, et al., 1999), upregulation of NG2 proteoglycan by oligodendrocyte precursor cells (Rhodes, et al., 2006), and cytokines associated with inflammation can also influence astrocyte extracellular matrix production (DiProspero et al., 1997).

It is important to consider inflammation within the CNS as a potential source of cytokines and other signaling molecules that can lead to upregulation of inhibitory molecules after an injury. Microglial cells from the CNS and activated macrophages from the periphery both respond to trauma in the brain and spinal cord, and this inflammatory response may contribute to secondary tissue damage after the primary insult (Blight, 1994; Fitch, et al., 1999). This cascade of secondary damage, progressive cavitation, and glial scarring was demonstrated in an in vivo model of inflammation using microinjection of zymosan, a non-toxic, specific phagocytic activator of the macrophage mannose receptor and the beta-glucan site of the CR3 integrin receptor (Fitch, et al., 1999). The resulting inflammation leads to significant axotomy and increases in astrocyte cavity size, while control injections of latex microspheres do not induce this damaging inflammatory cascade. Upregulation of inhibitory proteoglycans is also found at the interfaces between developing cavities and surrounding reactive astrocytes (Fitch and Silver, 1997a; MacLaren, 1996) further demonstrating the association of inflammation with upregulation of these molecules (Fitch, et al., 1999). The biological function of these inhibitory molecules when surrounding necrotic cavitation is unknown, but it is possible that these proteoglycans may be protective for the surrounding viable tissue to prevent further damage from the cavity progression that results from ongoing inflammatory processes (Fitch and Silver, 2007; Myer, et al., 2006).

Interestingly, inflammation within the CNS has been suggested to provide neuroprotective benefits during the healing process of the brain and spinal cord depending on the activation state of the cells (Klusman and Schwab, 1997; Lotan and Schwartz, 1994; Schwartz and Yoles, 2006). In addition to pro-inflammatory cytokines, macrophages can secrete factors that are growth promoting, such as NGF and NT-3 (Elkabes et al., 1996), thrombospondin (Chamak et al., 1994), and IL-1 (Giulian et al., 1994). Even zymosan, a trigger of robust inflammation, when placed in the vitreous chamber of the eye can stimulate regenerating optic nerve fibers (Leon et al., 2000) and when placed into the DRG before root crush appears essential to the success of DRG regeneration into the spinal cord when combined with modification of the extracellular matrix in the CNS compartment(Steinmetz, et al., 2005). Apparently intense inflammatory states created just outside of the CNS illustrate that in certain circumstances inflammation can promote enhanced regeneration by triggering a conditioning-like effect within the neuron (Calvo et al., 2005; Lund et al., 2002; McQuarrie and Jacob, 1991; Wu et al., 2007a). However, fulminant inflammation centrally tends to create cavities (Fitch, et al., 1999) and it is unclear whether further increasing inflammatory activity, even after attempting to modify immune cell activity, will lead to the desired functional regeneration within the brain and spinal cord (Kigerl et al., 2007; Kigerl et al., 2006; Lazarov-Spiegler et al., 1996; Schwartz and Yoles, 2006).

Modification of Extracellular Matrices to Enhance Regeneration

Strategies designed to enhance clinical outcomes and rehabilitation after spinal cord injury continues to be a topic of great interest, in particular because no treatment protocols with proven efficacy are currently in clinical use. While there was once great interest in the use of high-dose methylprednisolone for spinal cord injury (Bracken et al., 1990; Bracken et al., 1992), methodological problems with these studies and controversies surrounding subsequent trials have led to criticism and discontent with the routine use of this treatment strategy (Coleman et al., 2000; Hurlbert, 2001). Steroids were also previously thought to be helpful for patients with clinically significant head injuries until the recent randomized CRASH trial with over 10,000 enrolled patients demonstrated increased mortality when using methylprednisolone (Roberts et al., 2004). Modification of inflammatory processes and the resulting inhibitory extracellular matrices following trauma are likely to be important for improving clinical outcomes, but it appears that steroid treatment alone is not sufficient to provide reproducible and meaningful clinical recovery in human patients for spinal cord or brain injury (George et al., 1995; Pointillart et al., 2000; Prendergast et al., 1994; Roberts, et al., 2004; Tsutsumi et al., 2006).

Some other strategies for encouraging CNS neurons to regenerate are to increase local levels of growth factors in an effort to stimulate axonal growth. Genetically engineered fibroblasts secreting NGF can encourage axons to regenerate into the center of an injury when implanted in this region (Jones, et al., 2003b; Kawaja and Gage, 1991; Tuszynski et al., 1997), although this sprouting remains within the area of trophic support and does not lead to long distance functional regeneration. Intrathecally delivered NT-3, NGF, and GDNF can promote axonal regeneration across the dorsal root entry zone (Ramer et al., 2002; Ramer et al., 2000), and glial overexpression of NGF can stimulate regeneration of nociceptive axons from the dorsal roots which in some circumstances may lead to thermal hyperalgesia (Romero et al., 2000; Tang et al., 2004). Combination therapy such as treatment with NT-3 and cAMP together to stimulate neuronal cell bodies has been shown to allow regeneration beyond the lesion site of spinal injuries (Lu et al., 2004). The use of multiple simultaneous approaches to this complex problem may prove important when constructing future strategies for improving rehabilitation and functional recovery.

One important component to consider including in such combination approaches to therapy is modification of the inhibitory extracellular matrix molecules produced after injury. In vitro experiments have demonstrated that proteoglycans associated with reactive astrocytes can be modified to allow increased growth of axons by removing or preventing the production of sugar epitopes on the proteoglycan molecules (Bradbury, et al., 2002; Grimpe and Silver, 2004; McKeon, et al., 1995; McKeon, et al., 1991; Steinmetz, et al., 2005), and an in vivo transgenic model to express a chondroitin-sulfate proteoglycan degrading enzyme in astrocytes demonstrates a local efficacy for reductions in proteoglycans to enhance CNS axon growth after injury (Cafferty et al., 2007). Neutralizing a heparan/chondroitin sulphate proteoglycan expressed after brain injury with a blocking antibody will allow neurite outgrowth and prevent growth cone collapse in vitro (Bovolenta et al., 1997). Injured corticospinal fibers and uninjured serotonergic fibers increase sprouting after enzymatic treatment to remove sugar epitopes (Barritt et al., 2006), and in vivo studies have revealed remarkable long-distance regeneration of adult axons though CNS white matter tracts after enzymatic treatment to digest these proteoglycan side chains (Steinmetz, et al., 2005). The dorsal root entry zone is an important transition between the PNS and CNS where regenerating axons abruptly stop and fail to enter the CNS (Golding, et al., 1997; Kliot, et al., 1990; Perkins et al., 1980; Tower, 1931). This inhibitory interface can be overcome with a combination strategy that utilizes zymosan, a specific inflammatory activator of macrophages, and enzymatic digestion of inhibitory proteoglycans with chondroitinase ABC (Steinmetz, et al., 2005). Neither of these treatments alone were successful in vitro or in vivo, but together they led to robust regeneration of nerve fibers with electrophysiologically proven synapse formation. This is another example of a promising combination therapy, where a specific inflammatory environment is created outside the CNS using macrophage activators and the extracellular matrix in the CNS is modified to counteract the associated proteoglycan inhibitors.

Other recent therapeutic approaches have used the concept of combining different approaches to promote regeneration. Using an antibody to the NG2 proteoglycan along with a conditioning lesion to increase axon growth has recently been shown to enhance the regeneration of sensory fibers in the dorsal columns (Tan et al., 2006). A different combination strategy has resulted in functional regeneration of motor abilities when extracellular inhibitors are modified along with the use of PNS nerve bridges. Combining PNS nerve bridges with chondroitinase ABC to digest the sugar epitopes of chondroitin sulfate proteoglycans allows regenerating axons to bypass the spinal cord lesion and also allows axonal exodus from the conduit(Houle, et al., 2006). This results in reformation of functional synaptic connections with denervated interneurons and motor neurons below the level of the lesion. These animals had improved forelimb swing during locomotion, enhanced ability to use the forelimbs, and improved performance on a horizontal rope. All of these behavioral gains were diminished after transection of the PNS nerve bridge (Houle, et al., 2006). These findings emphasize that modification of inhibitory astrocyte molecules associated with certain inflammatory states after CNS injury are important considerations for combination strategies in spinal cord injury and rehabilitation.

Conclusion

Efforts are ongoing to further elucidate the critical components of CNS injury responses that prevent meaningful regeneration of injured nerve tracts. A key aspect of this response is the production of inhibitory extracellular matrix molecules via complex interactions between glial cells and inflammatory infiltrates, and the subsequent inhibitory effects on growing axons that leads to abortive regeneration and the formation of dystrophic neurons (see Figure 1). Recent work highlights the importance of a combination approach to therapy, and a successful strategy to improve patient rehabilitation after spinal injury will need to incorporate modification of inflammatory states, neutralization of inhibitory extracellular molecules, and stimulation of growth promoting factors by a variety of approaches.

Figure 1. Wound healing, secondary damage, and abortive regeneration in the central nervous system.

Figure 1

The lesion cavity of a central nervous system injury expands as inflammatory cells interact with the surrounding reactive astrocytes and other reactive glial cells. This region of glial scarring is associated with upregulation of inhibitory extracellular matrix molecules, such as proteoglycans, that are distributed in an increasing concentration gradient from the lesion penumbra to the lesion center. This intense inflammatory response leads to a cascade of secondary damage to axons initially spared from direct trauma, and demyelination of adjacent axons that are not readily re-myelinated by adult oligodendrocytes and precursor cells. The gradient of inhibitory molecules upregulated in the areas of intense inflammation provides an environment that is nonpermissive for regeneration, and dystrophic neurons develop the classically-described sterile end-balls with clubbed endings that are characteristic of abortive attempts at regeneration.

Acknowledgments

MTF received faculty funding support from the Brooks Scholars Program at the Wake Forest University School of Medicine. JS received research funding from NINDS / NS25713.

Footnotes

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

References

  1. Apostolova I, Irintchev A, Schachner M. Tenascin-R restricts posttraumatic remodeling of motoneuron innervation and functional recovery after spinal cord injury in adult mice. J Neurosci. 2006;26:7849–7859. doi: 10.1523/JNEUROSCI.1526-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ard MD, Schachner M, Rapp JT, Faissner A. Growth and degeneration of axons on astrocyte surfaces: effects on extracellular matrix and on later axonal growth. Glia. 1993;9:248–259. doi: 10.1002/glia.440090403. [DOI] [PubMed] [Google Scholar]
  3. Barrett CP, Guth L, Donati EJ, Krikorian JG. Astroglial reaction in the gray matter lumbar segments after midthoracic transection of the adult rat spinal cord. Exp Neurol. 1981;73:365–377. doi: 10.1016/0014-4886(81)90272-7. [DOI] [PubMed] [Google Scholar]
  4. Barritt AW, Davies M, Marchand F, Hartley R, Grist J, Yip P, McMahon SB, Bradbury EJ. Chondroitinase ABC promotes sprouting of intact and injured spinal systems after spinal cord injury. J Neurosci. 2006;26:10856–10867. doi: 10.1523/JNEUROSCI.2980-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bernstein JJ, Getz R, Jefferson M, Kelemen M. Astrocytes secrete basal lamina after hemisection of rat spinal cord. Brain Res. 1985;327:135–141. doi: 10.1016/0006-8993(85)91507-0. [DOI] [PubMed] [Google Scholar]
  6. Blight AR. Effects of silica on the outcome from experimental spinal cord injury: implication of macrophages in secondary tissue damage. Neuroscience. 1994;60:263–273. doi: 10.1016/0306-4522(94)90220-8. [DOI] [PubMed] [Google Scholar]
  7. Bovolenta P, Fernaud-Espinosa I, Mendez-Otero R, Nieto-Sampedro M. Neurite outgrowth inhibitor of gliotic brain tissue. Mode of action and cellular localization, studied with specific monoclonal antibodies. Eur J Neurosci. 1997;9:977–989. doi: 10.1111/j.1460-9568.1997.tb01448.x. [DOI] [PubMed] [Google Scholar]
  8. Bovolenta P, Wandosell F, Nieto-Sampedro M. Neurite outgrowth inhibitors associated with glial cells and glial cell lines. Neuroreport. 1993;5:345–348. doi: 10.1097/00001756-199312000-00042. [DOI] [PubMed] [Google Scholar]
  9. Bracken MB, Shepard MJ, Collins WF, Holford TR, Young W, Baskin DS, Eisenberg HM, Flamm E, Leo SL, Maroon J. A randomized, controlled trial of methylprednisolone or naloxone in the treatment of acute spinal-cord injury. Results of the Second National Acute Spinal Cord Injury Study. N Engl J Med. 1990;322:1405–1411. doi: 10.1056/NEJM199005173222001. al, e. [DOI] [PubMed] [Google Scholar]
  10. Bracken MB, Shepard MJ, Collins WJ, Holford TR, Baskin DS, Eisenberg HM, Flamm E, Leo SL, Maroon JC, Marshall LF. Methylprednisolone or naloxone treatment after acute spinal cord injury: 1-year follow-up data. Results of the second National Acute Spinal Cord Injury Study. J Neurosurg. 1992;76:23–31. doi: 10.3171/jns.1992.76.1.0023. al, e. [DOI] [PubMed] [Google Scholar]
  11. Bradbury EJ, Moon LD, Popat RJ, King VR, Bennett GS, Patel PN, Fawcett JW, McMahon SB. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature. 2002;416:636–640. doi: 10.1038/416636a. [DOI] [PubMed] [Google Scholar]
  12. Brodkey JA, Laywell ED, O’Brien TF, Faissner A, Stefansson K, Dorries HU, Schachner M, Steindler DA. Focal brain injury and upregulation of a developmentally regulated extracellular matrix protein. J Neurosurg. 1995;82:106–112. doi: 10.3171/jns.1995.82.1.0106. [DOI] [PubMed] [Google Scholar]
  13. Bundesen LQ, Scheel TA, Bregman BS, Kromer LF. Ephrin-B2 and EphB2 regulation of astrocyte-meningeal fibroblast interactions in response to spinal cord lesions in adult rats. J Neurosci. 2003;23:7789–7800. doi: 10.1523/JNEUROSCI.23-21-07789.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Busch SA, Silver J. The role of extracellular matrix in CNS regeneration. Curr Opin Neurobiol. 2007;17:120–127. doi: 10.1016/j.conb.2006.09.004. [DOI] [PubMed] [Google Scholar]
  15. Cafferty WB, Yang SH, Duffy PJ, Li S, Strittmatter SM. Functional axonal regeneration through astrocytic scar genetically modified to digest chondroitin sulfate proteoglycans. J Neurosci. 2007;27:2176–2185. doi: 10.1523/JNEUROSCI.5176-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Calvo CF, Amigou E, Desaymard C, Glowinski J. A pro- and an anti-inflammatory cytokine are synthesised in distinct brain macrophage cells during innate activation. J Neuroimmunol. 2005;170:21–30. doi: 10.1016/j.jneuroim.2005.08.005. [DOI] [PubMed] [Google Scholar]
  17. Canning DR, Hoke A, Malemud CJ, Silver J. A potent inhibitor of neurite outgrowth that predominates in the extracellular matrix of reactive astrocytes. Int. J. Devl. Neuroscience. 1996;14:153–175. doi: 10.1016/0736-5748(96)00004-4. [DOI] [PubMed] [Google Scholar]
  18. Canning DR, McKeon RJ, DeWitt DA, Perry G, Wujek JR, Frederickson RC, Silver J. beta-Amyloid of Alzheimer’s disease induces reactive gliosis that inhibits axonal outgrowth. Exp Neurol. 1993;124:289–298. doi: 10.1006/exnr.1993.1199. [DOI] [PubMed] [Google Scholar]
  19. Chamak B, Morandi V, Mallat M. Brain macrophages stimulate neurite growth and regeneration by secreting thrombospondin. J Neurosci Res. 1994;38:221–233. doi: 10.1002/jnr.490380213. [DOI] [PubMed] [Google Scholar]
  20. Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J Neurosci. 2000;20:6404–6412. doi: 10.1523/JNEUROSCI.20-17-06404.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chiang CS, Stalder A, Samimi A, Campbell IL. Reactive gliosis as a consequence of interleukin-6 expression in the brain: studies in transgenic mice. Dev Neurosci. 1994;16:212–221. doi: 10.1159/000112109. [DOI] [PubMed] [Google Scholar]
  22. Coleman WP, Benzel D, Cahill DW, Ducker T, Geisler F, Green B, Gropper MR, Goffin J, Madsen PW, 3rd, Maiman DJ, Ondra SL, Rosner M, Sasso RC, Trost GR, Zeidman S. A critical appraisal of the reporting of the National Acute Spinal Cord Injury Studies (II and III) of methylprednisolone in acute spinal cord injury. J Spinal Disord. 2000;13:185–199. doi: 10.1097/00002517-200006000-00001. [DOI] [PubMed] [Google Scholar]
  23. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:752–758. doi: 10.1038/nn1472. [DOI] [PubMed] [Google Scholar]
  24. David S, Aguayo AJ. Axonal elongation into peripheral nervous system “bridges” after central nervous system injury in adult rats. Science. 1981;214:931–933. doi: 10.1126/science.6171034. [DOI] [PubMed] [Google Scholar]
  25. Davies SJ, Fitch MT, Memberg SP, Hall AK, Raisman G, Silver J. Regeneration of adult axons in white matter tracts of the central nervous system. Nature. 1997;390:680–683. doi: 10.1038/37776. [DOI] [PubMed] [Google Scholar]
  26. Davies SJA, Goucher DR, Doller C, Silver J. Robust regeneration of adult sensory axons in degenerating white matter of the adult rat spinal cord. Journal of Neuroscience. 1999;19:5810–5822. doi: 10.1523/JNEUROSCI.19-14-05810.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. DiProspero NA, Meiners S, Geller HM. Inflammatory cytokines interact to modulate extracellular matrix and astrocytic support of neurite outgrowth. Experimental Neurology. 1997;148:628–639. doi: 10.1006/exnr.1997.6700. [DOI] [PubMed] [Google Scholar]
  28. Dou C-L, Levine JM. Inhibition of neurite growth by the NG2 chondroitin sulfate proteoglycan. J Neurosci. 1994;14:7616–7628. doi: 10.1523/JNEUROSCI.14-12-07616.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Elkabes S, DiCicco BE, Black IB. Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function. J Neurosci. 1996;16:2508–2521. doi: 10.1523/JNEUROSCI.16-08-02508.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fawcett JW. Overcoming inhibition in the damaged spinal cord. J Neurotrauma. 2006;23:371–383. doi: 10.1089/neu.2006.23.371. [DOI] [PubMed] [Google Scholar]
  31. Fitch MT, Doller C, Combs CK, Landreth GE, Silver J. Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma. J Neurosci. 1999;19:8182–8198. doi: 10.1523/JNEUROSCI.19-19-08182.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Fitch MT, Silver J. Activated macrophages and the blood-brain barrier: inflammation after CNS injury leads to increases in putative inhibitory molecules. Exp Neurol. 1997a;148:587–603. doi: 10.1006/exnr.1997.6701. [DOI] [PubMed] [Google Scholar]
  33. Fitch MT, Silver J. Glial cell extracellular matrix: boundaries for axon growth in development and regeneration. Cell Tissue Res. 1997b;290:379–384. doi: 10.1007/s004410050944. [DOI] [PubMed] [Google Scholar]
  34. Fitch MT, Silver J. Inflammation and the glial scar: Factors at the site of injury that influence regeneration in the central nervous system. In: Saunders NR, Dziegielewska KM, editors. Degeneration and regeneration in the nervous system. Harwood Academic Press; 2000. pp. 77–92. [Google Scholar]
  35. Fitch MT, Silver J. Astrocytes are Dynamic Participants in Central Nervous System Development and Injury Responses. In: Jessen KR, Richardson WD, editors. Glial Cell Development: Basic Principles and Clinical Relevance. Oxford University Press; London: 2001. pp. 263–269. [Google Scholar]
  36. Fitch MT, Silver J. Glial cells, inflammation and CNS trauma: Modulation of the inflammatory environment after injury can lead to long-distance regeration beyond the glial scar. In: Tuszynski MH, Kordower JH, editors. CNS Regeneration: Basic Science and Clinical Advances. Elsevier; 2007. In Press. [Google Scholar]
  37. Freund P, Schmidlin E, Wannier T, Bloch J, Mir A, Schwab ME, Rouiller EM. Nogo-A-specific antibody treatment enhances sprouting and functional recovery after cervical lesion in adult primates. Nat Med. 2006;12:790–792. doi: 10.1038/nm1436. [DOI] [PubMed] [Google Scholar]
  38. George ER, Scholten DJ, Buechler CM, Jordan-Tibbs J, Mattice C, Albrecht RM. Failure of methylprednisolone to improve the outcome of spinal cord injuries. Am Surg. 1995;61:659–663. discussion 663-654. [PubMed] [Google Scholar]
  39. Giulian D, Lachman LB. Interleukin-1 stimulation of astroglial proliferation after brain injury. Science. 1985;228:497–499. doi: 10.1126/science.3872478. [DOI] [PubMed] [Google Scholar]
  40. Giulian D, Li J, Li X, George J, Rutecki PA. The impact of microglia-derived cytokines upon gliosis in the CNS. Dev Neurosci. 1994;16:128–136. doi: 10.1159/000112099. [DOI] [PubMed] [Google Scholar]
  41. Golding J, Shewan D, Cohen J. Maturation of the mammalian dorsal root entry zone--from entry to no entry. Trends Neurosci. 1997;20:303–308. doi: 10.1016/s0166-2236(96)01044-2. [DOI] [PubMed] [Google Scholar]
  42. Goshgarian HG. The crossed phrenic phenomenon: a model for plasticity in the respiratory pathways following spinal cord injury. J Appl Physiol. 2003;94:795–810. doi: 10.1152/japplphysiol.00847.2002. [DOI] [PubMed] [Google Scholar]
  43. Grierson JP, Petroski RE, Ling DS, Geller HM. Astrocyte topography and tenascin cytotactin expression: correlation with the ability to support neuritic outgrowth. Brain Res Dev Brain Res. 1990;55:11–19. doi: 10.1016/0165-3806(90)90100-d. [DOI] [PubMed] [Google Scholar]
  44. Grimpe B, Silver J. A novel DNA enzyme reduces glycosaminoglycan chains in the glial scar and allows microtransplanted dorsal root ganglia axons to regenerate beyond lesions in the spinal cord. J Neurosci. 2004;24:1393–1397. doi: 10.1523/JNEUROSCI.4986-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Guenard V, Frisch G, Wood PM. Effects of axonal injury on astrocyte proliferation and morphology in Vitro: implications for astrogliosis. Exp Neurol. 1996;137:175–190. doi: 10.1006/exnr.1996.0017. [DOI] [PubMed] [Google Scholar]
  46. Guth L, Barrett CP, Donati EJ. Histological factors influencing the growth of axons into lesions of the mammalian spinal cord. Exp Brain Res Suppl. 1986;13:271–282. [Google Scholar]
  47. Guth L, Barrett CP, Donati EJ, Anderson FD, Smith MV, Lifson M. Essentiality of a specific cellular terrain for growth of axons into a spinal cord lesion. Exp Neurol. 1985;88:1–12. doi: 10.1016/0014-4886(85)90109-8. [DOI] [PubMed] [Google Scholar]
  48. Hagino S, Iseki K, Mori T, Zhang Y, Hikake T, Yokoya S, Takeuchi M, Hasimoto H, Kikuchi S, Wanaka A. Slit and glypican-1 mRNAs are coexpressed in the reactive astrocytes of the injured adult brain. Glia. 2003;42:130–138. doi: 10.1002/glia.10207. [DOI] [PubMed] [Google Scholar]
  49. Hama H, Kasuya Y, Sakurai T, Yamada G, Suzuki N, Masaki T, Goto K. Role of endothelin-1 in astrocyte responses after acute brain damage. J Neurosci Res. 1997;47:590–602. doi: 10.1002/(sici)1097-4547(19970315)47:6<590::aid-jnr4>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  50. Houle JD, Tom VJ, Mayes D, Wagoner G, Phillips N, Silver J. Combining an autologous peripheral nervous system “bridge” and matrix modification by chondroitinase allows robust, functional regeneration beyond a hemisection lesion of the adult rat spinal cord. J Neurosci. 2006;26:7405–7415. doi: 10.1523/JNEUROSCI.1166-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Hurlbert RJ. The role of steroids in acute spinal cord injury: an evidence-based analysis. Spine. 2001;26:S39–46. doi: 10.1097/00007632-200112151-00009. [DOI] [PubMed] [Google Scholar]
  52. Jones LL, Margolis RU, Tuszynski MH. The chondroitin sulfate proteoglycans neurocan, brevican, phosphacan, and versican are differentially regulated following spinal cord injury. Exp Neurol. 2003a;182:399–411. doi: 10.1016/s0014-4886(03)00087-6. [DOI] [PubMed] [Google Scholar]
  53. Jones LL, Sajed D, Tuszynski MH. Axonal regeneration through regions of chondroitin sulfate proteoglycan deposition after spinal cord injury: a balance of permissiveness and inhibition. J Neurosci. 2003b;23:9276–9288. doi: 10.1523/JNEUROSCI.23-28-09276.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kahn MA, Ellison JA, Speight GJ, de VJ. CNTF regulation of astrogliosis and the activation of microglia in the developing rat central nervous system. Brain Res. 1995;685:55–67. doi: 10.1016/0006-8993(95)00411-i. [DOI] [PubMed] [Google Scholar]
  55. Kawaja MD, Gage FH. Reactive astrocytes are substrates for the growth of adult CNS axons in the presence of elevated levels of nerve growth factor. Neuron. 1991;7:1019–1030. doi: 10.1016/0896-6273(91)90346-2. [DOI] [PubMed] [Google Scholar]
  56. Kerschensteiner M, Schwab ME, Lichtman JW, Misgeld T. In vivo imaging of axonal degeneration and regeneration in the injured spinal cord. Nat Med. 2005;11:572–577. doi: 10.1038/nm1229. [DOI] [PubMed] [Google Scholar]
  57. Kigerl KA, Lai W, Rivest S, Hart RP, Satoskar AR, Popovich PG. Toll-like receptor (TLR)-2 and TLR-4 regulate inflammation, gliosis, and myelin sparing after spinal cord injury. J Neurochem. 2007 doi: 10.1111/j.1471-4159.2007.04524.x. [DOI] [PubMed] [Google Scholar]
  58. Kigerl KA, McGaughy VM, Popovich PG. Comparative analysis of lesion development and intraspinal inflammation in four strains of mice following spinal contusion injury. J Comp Neurol. 2006;494:578–594. doi: 10.1002/cne.20827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kim SU, de Vellis J. Microglia in health and disease. J Neurosci Res. 2005;81:302–313. doi: 10.1002/jnr.20562. [DOI] [PubMed] [Google Scholar]
  60. Kliot M, Smith GM, Siegal JD, Silver J. Astrocyte-polymer implants promote regeneration of dorsal root fibers into the adult mammalian spinal cord. Exp Neurol. 1990;109:57–69. doi: 10.1016/s0014-4886(05)80008-1. [DOI] [PubMed] [Google Scholar]
  61. Klusman I, Schwab ME. Effects of pro-inflammatory cytokines in experimental spinal cord injury. Brain Res. 1997;762:173–184. doi: 10.1016/s0006-8993(97)00381-8. [DOI] [PubMed] [Google Scholar]
  62. Lazar DA, Fitch MT, Silver J, Kliot M. Cellular and molecular mechanisms mediating injury and recovery in the nervous system. In: Winn HR, editor. Youmans Neurological Surgery. WB Saunders; 2004. pp. 195–213. [Google Scholar]
  63. Lazarov-Spiegler O, Solomon AS, Zeev-Brann AB, Hirschberg DL, Lavie V, Schwartz M. Transplantation of activated macrophages overcomes central nervous system regrowth failure. FASEB J. 1996;10:1296–1302. doi: 10.1096/fasebj.10.11.8836043. [DOI] [PubMed] [Google Scholar]
  64. Le Gros Clark WE. The problem of neuronal regeneration in the central nervous system. II. The insertion of peripheral nerve stumps into the brain. J Anatomy. 1943;77:251–259. [PMC free article] [PubMed] [Google Scholar]
  65. Leon S, Yin Y, Nguyen J, Irwin N, Benowitz LI. Lens injury stimulates axon regeneration in the mature rat optic nerve. J Neurosci. 2000;20:4615–4626. doi: 10.1523/JNEUROSCI.20-12-04615.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Levine JM. Increased expression of the NG2 chondroitin-sulfate proteoglycan after brain injury. J Neurosci. 1994;14:4716–4730. doi: 10.1523/JNEUROSCI.14-08-04716.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Li Y, Raisman G. Schwann cells induce sprouting in motor and sensory axons in the adult rat spinal cord. J Neurosci. 1994;14:4050–4063. doi: 10.1523/JNEUROSCI.14-07-04050.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Li Y, Raisman G. Sprouts from cut corticospinal axons persist in the presence of astrocytic scarring in long-term lesions of the adult rat spinal cord. Exp Neurol. 1995;134:102–111. doi: 10.1006/exnr.1995.1041. [DOI] [PubMed] [Google Scholar]
  69. Liu BP, Cafferty WB, Budel SO, Strittmatter SM. Extracellular regulators of axonal growth in the adult central nervous system. Philos Trans R Soc Lond B Biol Sci. 2006;361:1593–1610. doi: 10.1098/rstb.2006.1891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Liu C-N, Chambers WW. Intraspinal sprouting of dorsal root axons. Arch. Neurol. Psychiat. 1958;79:46–61. [PubMed] [Google Scholar]
  71. Lotan M, Schwartz M. Cross talk between the immune system and the nervous system in response to injury: implications for regeneration. Faseb J. 1994;8:1026–1033. doi: 10.1096/fasebj.8.13.7926367. [DOI] [PubMed] [Google Scholar]
  72. Lu P, Yang H, Jones LL, Filbin MT, Tuszynski MH. Combinatorial therapy with neurotrophins and cAMP promotes axonal regeneration beyond sites of spinal cord injury. J Neurosci. 2004;24:6402–6409. doi: 10.1523/JNEUROSCI.1492-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lund LM, Machado VM, McQuarrie IG. Increased beta-actin and tubulin polymerization in regrowing axons: relationship to the conditioning lesion effect. Exp Neurol. 2002;178:306–312. doi: 10.1006/exnr.2002.8034. [DOI] [PubMed] [Google Scholar]
  74. MacLaren RE. Development and role of retinal glia in regeneration of ganglion cells following retinal injury. Br J Ophthalmol. 1996;80:458–464. doi: 10.1136/bjo.80.5.458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Massey JM, Hubscher CH, Wagoner MR, Decker JA, Amps J, Silver J, Onifer SM. Chondroitinase ABC digestion of the perineuronal net promotes functional collateral sprouting in the cuneate nucleus after cervical spinal cord injury. J Neurosci. 2006;26:4406–4414. doi: 10.1523/JNEUROSCI.5467-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. McGraw J, Hiebert GW, Steeves JD. Modulating astrogliosis after neurotrauma. J Neurosci Res. 2001;63:109–115. doi: 10.1002/1097-4547(20010115)63:2<109::AID-JNR1002>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
  77. McKeon RJ, Hoke A, Silver J. Injury-induced proteoglycans inhibit the potential for laminin-mediated axon growth on astrocytic scars. Exp Neurol. 1995;136:32–43. doi: 10.1006/exnr.1995.1081. [DOI] [PubMed] [Google Scholar]
  78. McKeon RJ, Schreiber RC, Rudge JS, Silver J. Reduction of neurite outgrowth in a model of glial scarring following CNS injury is correlated with the expression of inhibitory molecules on reactive astrocytes. J Neurosci. 1991;11:3398–3411. doi: 10.1523/JNEUROSCI.11-11-03398.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. McQuarrie IG, Jacob JM. Conditioning nerve crush accelerates cytoskeletal protein transport in sprouts that form after a subsequent crush. J Comp Neurol. 1991;305:139–147. doi: 10.1002/cne.903050113. [DOI] [PubMed] [Google Scholar]
  80. Misgeld T, Kerschensteiner M. In vivo imaging of the diseased nervous system. Nat Rev Neurosci. 2006;7:449–463. doi: 10.1038/nrn1905. [DOI] [PubMed] [Google Scholar]
  81. Misgeld T, Nikic I, Kerschensteiner M. In vivo imaging of single axons in the mouse spinal cord. Nat Protoc. 2007;2:263–268. doi: 10.1038/nprot.2007.24. [DOI] [PubMed] [Google Scholar]
  82. Murray M, Wang SD, Goldberger ME, Levitt P. Modification of astrocytes in the spinal cord following dorsal root or peripheral nerve lesions. Exp Neurol. 1990;110:248–257. doi: 10.1016/0014-4886(90)90036-r. [DOI] [PubMed] [Google Scholar]
  83. Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain. 2006;129:2761–2772. doi: 10.1093/brain/awl165. [DOI] [PubMed] [Google Scholar]
  84. Nishino A, Suzuki M, Ohtani H, Motohashi O, Umezawa K, Nagura H, Yoshimoto T. Thrombin may contribute to the pathophysiology of central nervous system injury. J Neurotrauma. 1993;10:167–179. doi: 10.1089/neu.1993.10.167. [DOI] [PubMed] [Google Scholar]
  85. Nishiyama A. Polydendrocytes: NG2 cells with many roles in development and repair of the CNS. Neuroscientist. 2007;13:62–76. doi: 10.1177/1073858406295586. [DOI] [PubMed] [Google Scholar]
  86. Pasterkamp RJ, Anderson PN, Verhaagen J. Peripheral nerve injury fails to induce growth of lesioned ascending dorsal column axons into spinal cord scar tissue expressing the axon repellent Semaphorin3A. Eur J Neurosci. 2001;13:457–471. doi: 10.1046/j.0953-816x.2000.01398.x. [DOI] [PubMed] [Google Scholar]
  87. Perkins S, Carlstedt T, Mizuno K, Aguayo AJ. Failure of regenerating dorsal root axons to regrow into the spinal cord. Canad J Neurol Sci. 1980;7:323. [Google Scholar]
  88. Pindzola RR, Doller C, Silver J. Putative inhibitory extracellular matrix molecules at the dorsal root entry zone of the spinal cord during development and after root and sciatic nerve lesions. Dev Biol. 1993;156:34–48. doi: 10.1006/dbio.1993.1057. [DOI] [PubMed] [Google Scholar]
  89. Pointillart V, Petitjean ME, Wiart L, Vital JM, Lassie P, Thicoipe M, Dabadie P. Pharmacological therapy of spinal cord injury during the acute phase. Spinal Cord. 2000;38:71–76. doi: 10.1038/sj.sc.3100962. [DOI] [PubMed] [Google Scholar]
  90. Prendergast MR, Saxe JM, Ledgerwood AM, Lucas CE, Lucas WF. Massive steroids do not reduce the zone of injury after penetrating spinal cord injury. J Trauma. 1994;37:576–579. doi: 10.1097/00005373-199410000-00009. discussion 579-580. [DOI] [PubMed] [Google Scholar]
  91. Ramer MS, Bishop T, Dockery P, Mobarak MS, O’Leary D, Fraher JP, Priestley JV, McMahon SB. Neurotrophin-3-mediated regeneration and recovery of proprioception following dorsal rhizotomy. Mol Cell Neurosci. 2002;19:239–249. doi: 10.1006/mcne.2001.1067. [DOI] [PubMed] [Google Scholar]
  92. Ramer MS, Priestley JV, McMahon SB. Functional regeneration of sensory axons into the adult spinal cord. Nature. 2000;403:312–316. doi: 10.1038/35002084. [DOI] [PubMed] [Google Scholar]
  93. Rhodes KE, Fawcett JW. Chondroitin sulphate proteoglycans: preventing plasticity or protecting the CNS? J Anat. 2004;204:33–48. doi: 10.1111/j.1469-7580.2004.00261.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rhodes KE, Raivich G, Fawcett JW. The injury response of oligodendrocyte precursor cells is induced by platelets, macrophages and inflammation-associated cytokines. Neuroscience. 2006;140:87–100. doi: 10.1016/j.neuroscience.2006.01.055. [DOI] [PubMed] [Google Scholar]
  95. Richardson PM, McGuinness UM, Aguayo AJ. Axons from CNS neurons regenerate into PNS grafts. Nature. 1980;284:264–265. doi: 10.1038/284264a0. [DOI] [PubMed] [Google Scholar]
  96. Roberts I, Yates D, Sandercock P, Farrell B, Wasserberg J, Lomas G, Cottingham R, Svoboda P, Brayley N, Mazairac G, Laloe V, Munoz-Sanchez A, Arango M, Hartzenberg B, Khamis H, Yutthakasemsunt S, Komolafe E, Olldashi F, Yadav Y, Murillo-Cabezas F, Shakur H, Edwards P. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): randomised placebo-controlled trial. Lancet. 2004;364:1321–1328. doi: 10.1016/S0140-6736(04)17188-2. [DOI] [PubMed] [Google Scholar]
  97. Romero MI, Rangappa N, Li L, Lightfoot E, Garry MG, Smith GM. Extensive sprouting of sensory afferents and hyperalgesia induced by conditional expression of nerve growth factor in the adult spinal cord. J Neurosci. 2000;20:4435–4445. doi: 10.1523/JNEUROSCI.20-12-04435.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Rostworowski M, Balasingam V, Chabot S, Owens T, Yong VW. Astrogliosis in the neonatal and adult murine brain post-trauma: elevation of inflammatory cytokines and the lack of requirement for endogenous interferon-gamma. J Neurosci. 1997;17:3664–3674. doi: 10.1523/JNEUROSCI.17-10-03664.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Schousboe A, Westergaarde N. Transport of neuroactive amino acids in astrocytes. In: Kettenmann H, Ransom BR, editors. Neuroglia. Oxford University Press; New York: 1995. pp. 246–258. [Google Scholar]
  100. Schwartz M, Yoles E. Immune-based therapy for spinal cord repair: autologous macrophages and beyond. J Neurotrauma. 2006;23:360–370. doi: 10.1089/neu.2006.23.360. [DOI] [PubMed] [Google Scholar]
  101. Silver J. Inhibitory molecules in development and regeneration. J Neurol. 1994;242:S22–S24. doi: 10.1007/BF00939236. [DOI] [PubMed] [Google Scholar]
  102. Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004;5:146–156. doi: 10.1038/nrn1326. [DOI] [PubMed] [Google Scholar]
  103. Smith-Thomas L, Fok-Seang J, Stevens J, Du JS, Muir E, Faissner A, Geller HM, Rogers JH, Fawcett JW. An inhibitor of neurite outgrowth produced by astrocytes. J Cell Sci. 1994;107:1687–1695. doi: 10.1242/jcs.107.6.1687. [DOI] [PubMed] [Google Scholar]
  104. Snow DM, Lemmon V, Carrino DA, Caplan AI, Silver J. Sulfated proteoglycans in astroglial barriers inhibit neurite outgrowth in vitro. Exp Neurol. 1990;109:111–130. doi: 10.1016/s0014-4886(05)80013-5. [DOI] [PubMed] [Google Scholar]
  105. Steinmetz MP, Horn KP, Tom VJ, Miller JH, Busch SA, Nair D, Silver DJ, Silver J. Chronic enhancement of the intrinsic growth capacity of sensory neurons combined with the degradation of inhibitory proteoglycans allows functional regeneration of sensory axons through the dorsal root entry zone in the mammalian spinal cord. J Neurosci. 2005;25:8066–8076. doi: 10.1523/JNEUROSCI.2111-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Steward O, Trimmer PA. Genetic influences on cellular reactions to CNS injury: the reactive response of astrocytes in denervated neuropil regions in mice carrying a mutation (Wld(S)) that causes delayed Wallerian degeneration. J Comp Neurol. 1997;380:70–81. doi: 10.1002/(sici)1096-9861(19970331)380:1<70::aid-cne5>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  107. Tan AM, Colletti M, Rorai AT, Skene JH, Levine JM. Antibodies against the NG2 proteoglycan promote the regeneration of sensory axons within the dorsal columns of the spinal cord. J Neurosci. 2006;26:4729–4739. doi: 10.1523/JNEUROSCI.3900-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Tang XQ, Cai J, Nelson KD, Peng XJ, Smith GM. Functional repair after dorsal root rhizotomy using nerve conduits and neurotrophic molecules. Eur J Neurosci. 2004;20:1211–1218. doi: 10.1111/j.1460-9568.2004.03595.x. [DOI] [PubMed] [Google Scholar]
  109. Taylor L, Jones L, Tuszynski MH, Blesch A. Neurotrophin-3 gradients established by lentiviral gene delivery promote short-distance axonal bridging beyond cellular grafts in the injured spinal cord. J Neurosci. 2006;26:9713–9721. doi: 10.1523/JNEUROSCI.0734-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Tom VJ, Doller CM, Malouf AT, Silver J. Astrocyte-associated fibronectin is critical for axonal regeneration in adult white matter. J Neurosci. 2004a;24:9282–9290. doi: 10.1523/JNEUROSCI.2120-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Tom VJ, Steinmetz MP, Miller JH, Doller CM, Silver J. Studies on the development and behavior of the dystrophic growth cone, the hallmark of regeneration failure, in an in vitro model of the glial scar and after spinal cord injury. J Neurosci. 2004b;24:6531–6539. doi: 10.1523/JNEUROSCI.0994-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Tower S. A search for trophic influence of posterior spinal roots on skeletal muscle, with a note on the nerve fibers found in the proximal stumps of the roots after excision of the root ganglia. Brain. 1931;54:99–110. [Google Scholar]
  113. Trotter J. NG2-positive cells in CNS function and the pathological role of antibodies against NG2 in demyelinating diseases. J Neurol Sci. 2005;233:37–42. doi: 10.1016/j.jns.2005.03.024. [DOI] [PubMed] [Google Scholar]
  114. Tsutsumi S, Ueta T, Shiba K, Yamamoto S, Takagishi K. Effects of the Second National Acute Spinal Cord Injury Study of high-dose methylprednisolone therapy on acute cervical spinal cord injury-results in spinal injuries center. Spine. 2006;31:2992–2996. doi: 10.1097/01.brs.0000250273.28483.5c. discussion 2997. [DOI] [PubMed] [Google Scholar]
  115. Tuszynski MH, Murai K, Blesch A, Grill R, Miller I. Functional characterization of NGF-secreting cell grafts to the acutely injured spinal cord. Cell Transplant. 1997;6:361–368. doi: 10.1177/096368979700600318. [DOI] [PubMed] [Google Scholar]
  116. Walz W. Role of glial cells in the regulation of the brain ion microenvironment. Prog Neurobiol. 1989;33:309–333. doi: 10.1016/0301-0082(89)90005-1. [DOI] [PubMed] [Google Scholar]
  117. Wilson HC, Scolding NJ, Raine CS. Co-expression of PDGF alpha receptor and NG2 by oligodendrocyte precursors in human CNS and multiple sclerosis lesions. J Neuroimmunol. 2006;176:162–173. doi: 10.1016/j.jneuroim.2006.04.014. [DOI] [PubMed] [Google Scholar]
  118. Windle WF, Chambers WW. Regeneration in the spinal cord of the cat and dog. J Comp Neurol. 1950;93:241–258. doi: 10.1002/cne.900930206. [DOI] [PubMed] [Google Scholar]
  119. Wolburg H, Risau W. Formation of the blood-brain-barrier. In: Kettenmann H, Ransom BR, editors. Neuroglia. Oxford University Press; New York: 1995. pp. 763–776. [Google Scholar]
  120. Wood P, Bunge RP. The biology of the oligodendrocyte. In: Norton WT, editor. Oligodendroglia. Plenum Press; New York: 1984. pp. 1–46. [Google Scholar]
  121. Wu D, Zhang Y, Bo X, Huang W, Xiao F, Zhang X, Miao T, Magoulas C, Subang MC, Richardson PM. Actions of neuropoietic cytokines and cyclic AMP in regenerative conditioning of rat primary sensory neurons. Exp Neurol. 2007a;204:66–76. doi: 10.1016/j.expneurol.2006.09.017. [DOI] [PubMed] [Google Scholar]
  122. Wu LJ, Vadakkan KI, Zhuo M. ATP-induced chemotaxis of microglial processes requires P2Y receptor-activated initiation of outward potassium currents. Glia. 2007b;55:810–821. doi: 10.1002/glia.20500. [DOI] [PubMed] [Google Scholar]
  123. Yiu G, He Z. Glial inhibition of CNS axon regeneration. Nat Rev Neurosci. 2006;7:617–627. doi: 10.1038/nrn1956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Zhang H, Uchimura K, Kadomatsu K. Brain keratan sulfate and glial scar formation. Ann N Y Acad Sci. 2006;1086:81–90. doi: 10.1196/annals.1377.014. [DOI] [PubMed] [Google Scholar]

RESOURCES