Open access peer-reviewed chapter

Bridging Defects in Chronic Spinal Cord Injury Using Peripheral Nerve Grafts: From Basic Science to Clinical Experience

Written By

Sherif M. Amr

Submitted: 21 October 2015 Reviewed: 12 May 2016 Published: 17 August 2016

DOI: 10.5772/64211

From the Edited Volume

Recovery of Motor Function Following Spinal Cord Injury

Edited by Heidi Fuller and Monte Gates

Chapter metrics overview

1,699 Chapter Downloads

View Full Metrics

Abstract

Nerve grafting of the injured spinal cord should pursue a sixfold attack: lysing the fibrosis/gliosis to an extent that allows settling of the basal lamina preventing meanwhile collapse of the neural tissue matrix; supplying the tissue matrix with a suitable scaffold, on which the basal lamina can settle; basal lamina synthesis; seeding the basal lamina with cell adhesion molecules; providing the axonal growth cone with neurite outgrowth promoting factors that allow its distal progression; supplying the axonal growth cone with neurotrophic factors that power its continued growth. In addition to this, the intrinsic properties of the neurons should be stimulated, possibly through modulating the function of astrocytes by heparin, aspirin and other factors. Nerve side grafting of the cord increases the incidence of nerve regeneration by applying additional grafts extending from the side of the donor end of the cord to the side of the recipient end. Also, it allows the surgeon to enhance regeneration through a partially regenerated cord. During surgery, after establishment of CSF circulation, a long-lasting indwelling catheter has to be inserted for postoperative drug and cell delivery. This allows for continual lysis of the gliosis by chondroitinase ABC, sialidase and other factors.

Keywords

  • spinal cord injury
  • nerve grafting
  • indwelling intrathecal catheter implantation
  • lysis of the gliosis
  • chondroitinase ABC

1. Introduction

Since 1903, when Tello and Cajal demonstrated that the central nervous system (CNS) could regenerate [1, 2], experimental neuroscience has advanced our knowledge repairing the injured spinal cord. Several cellular transplantation strategies have been recommended with some clinical success [36]. Clinically, however, the injured spinal cord is usually extensively gliotic, cystic, even disrupted, necessitating bridging the injury zone first before contemplating cellular transplantation. Placing peripheral nerve grafts to bridge the injury zone has been successful experimentally [713], yet only anecdotal clinical evidence supports it [14, 15]. In a review article on bridging spinal cord injuries, Fawcett [16] commented on this disparity between experimental and clinical neuroscience: ‘Sadly, we have yet to achieve a treatment that is licensed for this purpose in human patients’. The aim of this review is to enable clinicians to put the findings made by neuroscientists into clinical practice and to provide neuroscientists with upcoming ideas investigating the clinical issues physicians face.

Advertisement

2. Changing concepts of nerve grafting

2.1. Basic concepts of nerve grafting

Autogenous nerve grafting is the standard for repair of irreducible nerve gaps [17]. The basic principles of nerve grafting include the following (Figure 1):

Figure 1.

The basic principles of conventional end-to-end grafting include trimming both proximal and distal nerve ends up to healthy nerve fascicles; avoiding any tension at the repair site, avoiding any shearing stress at the repair site, fascicular grafting, end-to-end grafting suturing fascicles at proximal nerve ends to their counterparts at distal nerve ends after grouping them topographically, using small caliber sutures (9/0 or 10/0 sutures), and healthy vascular bed.

  1. - trimming both proximal and distal nerve ends up to healthy nerve fascicles;

  2. - avoiding any tension at the repair site, for even minimal tension, can end up with fibrosis, hampering progression of regeneration;

  3. - avoiding any shearing stress at the repair site because this incites an inflammatory reaction ending up with fibrosis and hampering progression of regeneration;

  4. - fascicular grafting because autogenous nerve grafts derive their nutrition from the extracellular matrix; using large diameter grafts instead of small diameter fascicles can produce central necrosis of the graft;

  5. - end-to-end grafting suturing fascicles at proximal nerve ends to their counterparts at distal nerve ends after grouping them topographically, in order to avoid aberrant nerve sprouting;

  6. - using small caliber sutures (9/0 or 10/0 sutures) because large caliber sutures may produce fibrosis;

  7. - healthy vascular bed because a fibrotic bed may prevent progression of regeneration through the grafts.

In the absence of a proximal nerve end, such as in brachial plexus avulsions, nerve transfer (neurotisation) refers to using an expendable nearby donor nerve as a substitute, grafting it to the original recipient. The principles of nerve transfer include:

  1. - donor nerve of high axonal load,

  2. - single donor to single recipient to prevent cocontractions.

Autogenous grafts act as immunogenically inert scaffolds, providing appropriate neurotrophic factors and viable Schwann cells for axonal regeneration [17].

2.2. Molecular aspects of peripheral nerve regeneration

Advances in the understanding of molecular pathways and their physiological role have provided us with new insights as to the mechanism of axonal (peripheral nerve) regeneration [18, 19].

Fibrous tissue and chondroitin sulphate proteoglycans secreted by astrocytes provide the necessary scaffold for settling of the basal lamina and subsequent basement membrane synthesis. Neurite outgrowth promoting factors are basement membrane-related extracellular matrix proteins (such as laminin (LN), fibronectin (FN), heparin sulphate proteoglycans (HSP) and tenascin), which pave the proper path by supplying orientation and adhesiveness for axons. Neurotrophic factors are specific trophic agents that power peripheral nerve regeneration. They include (a) the neurotrophins (nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and neurotrophin-4/5(NT-4/5)); (b) the neurokines (ciliary neurotrophic factor (CNTF) and leukaemia inhibitory factor (LIF)); (c) the transforming growth factor (TGF)-b family (TGF-b1, TGF-b2, TGF-b3, glial cell line¦derived neurotrophic factor (GDNF)). Schwann cells secrete (a) cell adhesion molecules (CAMs), such as N-CAM, Ng-CAM/L1, N-cadherin and L2/HNK-1; (b) produce basement membrane that contains many extracellular matrix proteins, such as laminin (LN), fibronectin (FN), heparin sulphate proteoglycans (HSP) and tenascin; (c) secrete neurotrophic factors and their receptors. In addition to this, axonal regeneration is determined by trophic factors from activated perineural glial cells (astrocytes), trophic factors from efferent axons by anterograde transport, gene-induced trophic factors by intracrine or autocrine transport, trophic factors from retrograde target cell support, trophic factors from retroaxonal transport and trophic factors from recruited macrophages (secretory products, cytokines). Activated mesenchymal cells contribute to repair and vascularisation (Figure 2).

Figure 2.

Molecular aspects of axonal regeneration. (I) Neurite outgrowth promoting factors [such as laminin (LN), fibronectin (FN), heparin sulphate proteoglycans (HSP) and tenascin] pave the proper path by supplying orientation and adhesiveness for axons. (II) Neurotrophic factors are the specific trophic agents that power peripheral nerve regeneration. They include (a) neurotrophins (NGF, BDNF, NT-4/5); (b) neurokines (CNTF, LIF); (c) (TGF)-β family (TGF-β1, TGF-β2, TGF-β3, GDNF). (III) Schwann cells secrete (a) cell adhesion molecules (CAMs), such as N-CAM, Ng-CAM/L1, N-cadherin, andL2/HNK-1; (b) basement membrane that contains many extracellular matrix proteins, such as laminin (LN), fibronectin (FN), heparin sulphate proteoglycans (HSP) and tenascin; (c) neurotrophic factors and their receptors. In addition to this, axonal regeneration is determined by trophic factors from activated perineural glial cells (astrocytes) (IV), trophic factors from efferents by anterograde transport (V), gene-induced trophic factors by intracrine (VI) or autocrine (VII) transport, trophic factors from retrograde target cell support (VIII), trophic factors from retroaxonal transport (IX), trophic factors from recruited macrophages (secretory products X, cytokines XI). Activated mesenchymal cells contribute to repair and vascularisation (XII). Among other molecules, heparin and aspirin modulate astrocytic function stimulating them to secrete axonal trophic factors (IV).

Based on the previous considerations, axonal sprouting and nerve grafting are based on a sixfold attack (Figure 3):

  1. – lysing the fibrosis/gliosis in the injury zone to an extent that allows settling of the basal lamina preventing meanwhile collapse of the neural tissue matrix; or excision of the fibrotic segment and replacing it with nerve grafts;

  2. – supplying the tissue matrix with a suitable scaffold, on which the basal lamina can settle,

    Figure 3.

    The sixfold attack: lysing the fibrosis/gliosis to an extent that allows settling of the basal lamina preventing meanwhile collapse of the neural tissue matrix; supplying the tissue matrix with a suitable scaffold, on which the basal lamina can settle; basal lamina synthesis; seeding the basal lamina with cell adhesion molecules; providing the axonal growth cone with neurite outgrowth promoting factors (FGF) that allow its distal progression; supplying the axonal growth cone with neurotrophic factors that allow its continued growth.

  3. – basal lamina synthesis;

  4. – seeding the basal lamina with cell adhesion molecules;

  5. – providing the axonal growth cone with neurite outgrowth promoting factors that allow its distal progression;

  6. – supplying the axonal growth cone with neurotrophic factors that power its continued growth.

2.3. Changing concepts of nerve grafting: side grafting

The previous conditions prevailing, if the side of a motor nerve is injured, the axonal growth cone may be enticed to grow off motor nerve side to the injured end of another motor nerve, the so-called recipient end to donor side coaptation. Described independently by Balance and Harris over a century ago (in 1903), interest in end-to-side coaptation has been rekindled by Viterbo et al. [17]. In its essence, it involves grafting donor side to recipient end after stimulating donor side collateral sprouting by mechanical trauma or axotomy (Figure 4(a)). An indirect application of it is increasing the incidence of nerve regeneration after conventional end-to-end grafting by applying additional grafts extending from the side of the donor end to the side of the recipient end [20] (Figure 4(b)). In nerve transfer, the latter technique allows the surgeon to use a single high axonal load donor for multiple recipients without producing cocontractions (e.g., major brachial plexus root to several peripheral nerves and caudal cord to cauda equina) [20] (Figure 4(c)). Also, partially regenerated nerves cannot be surgically cut and nerve grafted leading to loss of already regained function; the latter technique allows the surgeon to enhance regeneration through them (Figure 4(d)).

Figure 4.

(a) End-to-side grafting involves grafting donor side to recipient end after stimulating donor side collateral sprouting by mechanical trauma or axotomy. (b) An indirect application of side grafting is increasing the incidence of nerve regeneration after conventional end-to-end grafting by applying additional grafts extending from the side of the donor end to the side of the recipient end. (c) Side grafting allows the surgeon to use a single high axonal load donor for multiple recipients without producing cocontractions. (d) Partially regenerated nerves cannot be surgically cut and nerve grafted sacrifying already regained function; side grafting allows the surgeon to enhance regeneration through them.

2.4. Mathematical modelling and channel-carrying capacity applied to nerve grafting: necessary concepts for subsequent computer-assisted fabrication of artificial nerve grafts

Can we manipulate the molecular mechanisms of the sixfold attack to increase neurite outgrowth into the side grafts? Manipulating molecular mechanisms is based on the sensitivity of the axonal growth cone to spatial molecular concentration gradients [21, 22]. In a study by Rosoff et al. [21], axonal growth has been shown to be enhanced by a steep nerve growth factor (NGF) spatial concentration gradient. There is a narrow range, however, between the lowest NGF concentration necessary for axonal growth stimulation and the highest NCF concentration beyond which axonal growth is competitively blocked. As the lower and upper limits of the concentration gradient should fall within this narrow range, the maximal distance for axonal growth cone progression guided by that gradient would be far less than the length of the neural defect. Utilising synthetic nerve graft scaffolds and observing the sixfold attack, axonal growth can be hypothetically made to bridge the whole length of the neural gap by seeding the scaffolds with multiple NGF spatial concentration gradients [22]. Neurite outgrowth has been modelled as a non-linear partial differential equation, that is solved by an iterative mathematical process suitable for numerical analysis and for subsequent computer-assisted fabrication of artificial nerve grafts [22]. By diffusion, these NGF spatial concentration gradients might also enhance axonal growth within the adjacent natural nerve side grafts.

Alternatively, and also to increase neurite outgrowth into the side grafts and decrease aberrant neural sprouting, multiple microspheres embedding chemical attractive and repulsive cues and placed along nerve side grafts may be used to guide axonal growth [23]. Preliminary experiments conducted with embryonic rat hippocampal neurons and calcium alginate microspheres have been encouraging [24]. A mathematical model has been developed based on the diffusion gradient of the implanted microspheres; a genetic algorithm has been used to study its proper spatial implementation [23].

A more accurate mathematical model for axonal growth cone progression has been provided based on sensory pinch test data [25]. Disadvantages of this model, however, include the assumptions that the initial delay is the major cause of variability, and that delay to scarring of the neural bed lies within the initial delay and that the regeneration rate is linear (constant).

Can we quantify the molecular mechanisms of the sixfold attack so that nearly 100% of all axons sprouting from the proximal spinal cord reach the distal spinal cord through the side grafts and simultaneously minimise the probability of aberrant neural sprouting to nearly 0%? Unless incorporated in information theory, which is a theory based on mathematical probability [26, 27], the mathematical models mentioned above do not provide a numerical solution for this problem. This is imperative, however, for subsequent computer-assisted fabrication of artificial nerve grafts. The Shannon-Hartley channel-carrying capacity principle, a central concept in information theory, refers to the intrinsic property of any information channel to accept all information from the donor and transmit it noiseless to the recipient. Applied to nerve grafting, the channel-carrying capacity of a nerve graft scaffold is its ability to transmit all axons sprouting from the proximal cord to the distal cord and simultaneously minimise the probability of aberrant neural sprouting.

Advertisement

3. Peripheral nerve grafting of the injured spinal cord as an application of the concept of side grafting and the sixfold attack

3.1. Technical aspects of peripheral nerve grafting and repair of the injured spinal cord

An indirect application of side grafting, as mentioned previously, is, first, increasing the incidence of nerve regeneration after conventional end-to-end grafting by applying additional grafts extending from the side of the donor end to the side of the recipient end; and, second, preserving partially regenerated nerves which cannot be surgically cut and nerve grafted leading to loss of already regained function [20]. Both of these apply to the spinal cord; compared to its high axonal load, the cross-sectional area of the spinal cord is too small for efficient end-to-end grafting. In addition, some kind of regeneration may have occurred in a contused cord in a completely paraplegic patient; this may take the form of less than Grade 3 motor power improvement, which, according to ASIA standards, is not enough to be considered motor or neurological level progression [28]. Third, side grafting produces less trauma to the spinal cord. In fact, the glial tissue secreted by astrocytes provides the necessary supporting tissue for axons and neurons [2, 29]. In side grafting, fine pial incisions are used. This process is far less traumatic than freshening of the cord ends during end-to-end grafting, a procedure which would lead to excessive glial tissue secretion and subsequent blocking of regeneration.

3.2. Donor nerves

Clinically, the sural nerve is the most commonly used donor nerve, other suitable donor nerves include the medial and lateral cutaneous nerves of the forearm, dorsal cutaneous branch of the ulnar nerve, superficial and deep peroneal nerves, intercostal nerves, and the posterior and lateral cutaneous nerves of the thigh [17].

Pre-degenerated (segments of nerve cut and left in situ for 7–10 days prior to harvesting) peripheral nerves are infiltrated by regenerating axons to a greater extent (both in number and distance) than freshly cut and harvested nerves [30]. The use of pre-degenerated nerves has been contested by other authors, however [31].

3.3. Experimental applications of side grafting

Based on the work of David and Aguayo [5, 7], numerous studies have confirmed axonal outgrowth after nerve grafting of the injured spinal cord [30, 3238]; axons growing within peripheral nerve grafts have not only retained their physiological properties [39] but have synapsed with neurons near the point of central nervous system re-entry as well [40].

3.4. Re-evaluation of the use of peripheral nerve grafts to bridge spinal cord defects

Nerve grafts supply the injured spinal cord with five factors of the sixfold attack: a suitable scaffold, on which the basal lamina can settle; basal laminae; cell adhesion molecules; neurite outgrowth promoting factors; and neurotrophic factors. Nevertheless, the use of peripheral nerve grafts has been challenged [5, 16, 35, 41, 42]. According to experimental observation, damaged spinal cord axons might grow from the cranial cord into peripheral nerve grafts but would not leave them to enter the caudal cord [5]. Schwann cells might even promote gliosis [16, 41].

Figure 5.

(a) Excessive fibrosis in the presence of neurotophic factors and adequate neuron growth potential leads to hypertrophy of both donor and recipient ends of the damaged neural tissue (excessive neuroma formation; dystrophic growth cones). (b) Excessive fibrosis in the absence of neurotophic factors and adequate neuron growth potential leads to atrophy of both donor and recipient ends of the damaged neural tissue (atrophic neuroma formation; atrophic growth cones).

It might be assumed that failure of growth cone progression is due to missing of the sixth factor in the sixfold attack (lysing the fibrosis/gliosis to an extent that allows settling of the basal lamina preventing meanwhile collapse of the neural tissue matrix). However, failure of axons to regenerate following peripheral nerve grafting may also result from intrinsic properties of the neurons and the absence of neurotrophic factors [4346]. These considerations help us assess neural tissue during surgery. Excessive fibrosis in the presence of neurotophic factors and adequate neuron growth potential leads to hypertrophy of both donor and recipient ends of the damaged neural tissue (excessive neuroma formation and dystrophic growth cones) [47, 48] (Figure 5(a)), whereas excessive fibrosis in the absence of neurotophic factors and adequate neuron growth potential leads to atrophy of both donor and recipient ends of the damaged neural tissue (atrophic neuroma formation; atrophic growth cones) (Figure 5(b)). In conclusion, in addition to the sixfold attack, the intrinsic properties of the neurons should be stimulated to produce neurites.

Advertisement

4. Lysing the gliosis in conjunction with nerve grafting

4.1. The gliosis: blocking of regeneration mechanically and through the activation of RhoA

Injured axons encounter a series of inhibitory factors that are non-permissive for growth [4, 29, 41, 49]. These include myelin inhibitors (Nogo-A ,MAG108 (myelin-associated glycoprotein) and OMgp109 (oligodendrocyte myelin glycoprotein)); chondroitin sulphate proteoglycans (neurocan, versican, aggrecan, brevican, phosphacan and NG2); and semaphorins and ephrins. Upregulated in response to spinal cord injury [5052], they act not only by mechanical blocking of neural regeneration but by inhibiting axon outgrowth neuronal receptors and subsequent activation of signalling pathways known to be involved in the activation of RhoA and the rise in intracellular calcium.

4.2. Lysing the gliosis by chondroitinase ABC

Chondroitinase ABC cleaves the inhibitory chondroitin sulphate glycosaminoglycan chains from the core protein, reducing the inhibition by chondroitin sulphate proteoglycans [53, 54]. Houle et al. [55] have demonstrated CNS axons regenerating through a peripheral nerve graft and entering the caudal spinal cord following chondroitinase ABC treatment.

4.2.1. Chondroitinase ABC in combination with growth factors and scaffolds

Combined with glial-derived neurotrophic factor (GDNF) delivery, chondroitinase ABC promotes axon extension through peripheral nerve bridges [50, 56]. Combined with growth factors, chondroitinase ABC enhances the activation and oligodendrocyte differentiation of endogenous precursor cells after spinal cord injury and attenuates astrogliosis. When added to polycaprolactone or poly (acrylonitrile)/poly(vinyl chloride) (PAN/PVC) scaffolds, chondroitinase ABC allows regenerating axons to exit the distal end of the scaffold and continue on to distal targets [5760].

4.2.2. Chondroitinase ABC in combination with cell therapies

Chondroitinase ABC has improved recovery of function in synergy with mesenchymal stromal cells [D43], or a Schwann cell bridge and olfactory ensheathing cells or in combination with transplanted neural precursor cells and a growth factor (GF) cocktail containing EGF, FGF2 and platelet-derived growth factor (PDGF)-AA [61, 62].

4.2.3. Thermostabilisation, delivery, dosage and complications of chondroitinase ABC: chondroitinase ABC might be a weak enzyme

Thermostabilisation of chondroitinase ABC with the sugar trehalose can reduce its temperature-dependent loss of activity [50, 63]. Delivery of chondroitinase ABC is predominantly intrathecal using osmotic minipumps [30, 55, 64]. Nevertheless, the enzyme can also be loaded into lipid microtubes or possibly poly (lactic-co-glycolic acid) (PLGA), providing a means for its gradual release over 1–2 weeks [65]. It can also be loaded into fibrin gel scaffolds before injecting it intrathecally; this ensures its continuous release for at least 3 weeks [66].

The effect of chondroitinase ABC is dose-dependent [67]. Injected intrathecally in acute injuries at a low dose (1 or 5 IUs), chondroitinase ABC may enhance axonal progression; injected at a high dose (50 IU), it may produce subarachnoid haemorrhage. In subacute or chronic injuries, low-dose injection produces no or limited functional recovery, whilst high-dose injection (50 or 100 IUs) produce lysis of the gliosis [68, 69].

Single-dose chondroitinase ABC is not considered enough. Therefore, multiple single injections at 0, 1, 2 and 4 weeks have been recommended [70]. Daily injections for 2 weeks at 0.06 Units per dose have also been recommended [71]. Four weeks of treatment have promoted recovery more than 2 weeks [72]. One way to ensure the continued release of chondroitinase ABC is neuron transfection with a vector containing the gene encoding chondroitinase ABC [73]. Another way is loading scaffolds with chondroitinase ABC.

Although chondroitinase ABC allows substantial structural plasticity in the spinal cord, it is not sufficient to enhance locomotor recovery unless combined with neural precursor cell transplantation and in vivo infusion of growth factors [74]. In a rat spinal cord injury model, Wilems et al. [62] have used fibrin scaffolds loaded with neurotrophic factors (item I), anti-inhibitory molecules (item II) and encapsulated embryonic stem-cell-derived progenitor motor neurons (pMNs) (item III). Fibrin scaffolds containing items I and II but not item III have had lower chondroitin sulphate proteoglycan levels compared to scaffolds containing items II and III. This shows the importance of combining neurotrophic factors with chondroitinase ABC and cellular transplants. Scaffolds containing item III, but not item II, have shown differentiation into neuronal cell types, axonal extension and the ability to integrate into host tissue. However, the combination of items II and III have led to reduced cell survival and increased macrophage infiltration. This shows that cellular transplants not only claim priority over chondroitinase ABC, but that chondroitinase ABC may be a weak enzyme as well. Because of this fact, a combination treatment of zymosan and chondroitinase ABC has been recommended [75]. Lastly, adipose-derived stem cell transplantation has been found to produce the same effect as chondroitinase ABC administration [76]. Thus, chondroitinase ABC might be a weak enzyme.

4.3. Sialidase as an alternative

In a rat model of spinal cord contusion injury the effects of sialidase (Vibrio cholerae) and chondroitinase ABC (ChABC, Proteus vulgaris) have been tested [77]. Immunohistochemistry has revealed that infused sialidase has acted robustly throughout the spinal cord grey and white matter, whereas ChABC activity has been more intense superficially. Sialidase treatment alone has resulted in improved behavioural and anatomical outcomes.

4.4. Anti-Nogo

Blocking myelin-associated inhibitors with Nogo-A monoclonal antibodies or with Nogo-receptor competitive agonist peptide, NEP1-40 has been shown to increase axonal regeneration [50]. Combination therapies, such as cross-linking the Nogo-66 receptor antibody into a hyaluronic acid hydrogel [50], a combination of methylprednisolone and NEP1-40 and Nogo-receptor vaccination combined with neural stem cell transplantation have improved neural fibre regeneration.

4.5. Rho inhibition

Many of the inhibitory signals described above (ephrins, Nogo) converge on the intracellular molecule Rho-A, which is a key mediator of actin depolymerisation and hence inhibition of axonal elongation. Blocking Rho-A with Rho inhibitor ‘cethrin’ might overcome its effect. A synthetic membrane-permeable peptide mimetic of the protein tyrosine phosphatase σ (PTPσ), wedge domain can bind to PTPσ and relieve chondroitin sulphate proteoglycan-mediated inhibition [78].

4.6. Reversing the inhibition of phosphoinositide 3-kinase (PI3K) by cell permeable phosphopeptide (PI3Kpep)

Phosphoinositide 3-kinase (PI3K) is a lipid kinase activated by axon growth promoting signals. Chondroitin sulphate proteoglycans inhibit phosphoinositide 3-kinase signalling in axons and growth cones, an effect that can be reversed by cell permeable phosphopeptide (PI3Kpep). The latter acts by R-Ras-PI3K signalling [79].

4.7. Rolipram

Increased intracellular levels of cyclic adenosine monophosphate (cAMP) and protein kinase A have been associated with CNS ability to overcome the gliosis [50]. Rolipram, a phosphodiesterase4 inhibitor, can increase intracellular cAMP levels [50].

4.8. Improving blood vessel formation

Improving blood vessel formation might reduce cell death and promote angiogenesis within the injury zone. Neural stem cells modified to express vascular endothelial growth factor have improved white matter sparing following thoracic contusion spinal cord injury [50]. Biomaterial poly-lactic-co-glycolic acid (PLGA) scaffolds loaded with neural stem cells and endothelial cells have shown increased vessel and neurofilament density at the injury centre [50].

Advertisement

5. Modulating astrocyte function enhances the intrinsic properties of neurons to stimulate neurite outgrowth into peripheral nerve grafts

In addition to the sixfold attack, the intrinsic properties of the neurons have to be stimulated to produce neurites. This can take place by modulating astrocyte function (Figure 2).

5.1. Endogenous inhibitors of axonal regeneration (intrinsic properties of neurons)

Endogenous inhibitors of axonal regeneration include the molecule phosphatase and tensin homologue (PTEN), loss of neuronal cAMP and deactivation/activation of certain transcription factors [46].

The molecule phosphatase and tensin homologue (PTEN) on chromosome 10 is a tumour suppressor. Its deletion has been shown to increase post-embryonic neural regeneration after injury. Its inhibition–mediated regeneration is partly mediated by the inhibitor of the mechanistic target of rapamycin (mTOR) pathway; it is also mediated by glycogen synthesis kinase GSK-3β.

When levels of cAMP at the growth cone are high, the effect on the growth cone is chemoattraction, whereas when they are low, the effect is chemorepulsion.

Certain transcription factors are positive regulators of axonal growth (e.g., members of the Krüppel-like factors (KLFs) present in retinal ganglion cells (RGCs); STAT3 a transcription factor, part of the JAK-STAT signaling pathway; members of the Jun and Fos families, components of the transcription factor AP-1 and ATF3). Other transcription factors are negative regulators of axonal growth. Nuclear factor IL-3 (NFIL3) represses CREB-mediated transcription and expression of regeneration-associated genes such as arginase and GAP-43.

It follows that combatting endogenous inhibitors of axonal regeneration include the following:

  1. – Inactivation of GSK-3β by neurotrophins. This increases collapsin response mediator protein-2 (CRMP-2) stabilisation of microtubules and increases axon elongation in developing neurites.

  2. – Local administration of taxol. Microtubules and actin microfilaments are critical for regeneration [80]. They potentiate the effect of GAP-43. Thus, local administration of taxol, a microtubule-stabilising agent, increases neurite outgrowth [81].

  3. – Elevating cAMP levels by local injection of a phosphodiesterase inhibitor. This improves axonal regeneration.

  4. – Conditioning lesions. Conditioning lesions [46] are based on the observation that double level nerve lesions regenerate better than single level nerve lesions. Thus sciatic nerve transection prior to a spinal cord lesion improves regeneration within the injured spinal cord.

  5. – Cell adhesion molecules. Their synthesis is increased after peripheral nerve injury but not after CNS injury [82, 83]. Expression of cell adhesion molecules by neurons can be induced by virally mediated vectors or by injecting them into the CNS injury site [84, 85].

Many of these functions can be activated by modulating astrocyte function.

5.2. Astrocyte trophic effects on neurite outgrowth and neurogenesis

Astrocytes release a variety of trophic factors [8688]. These trophic factors include nerve growth factor, basic fibroblast growth factor, transforming growth factor-β, platelet-derived growth factor, brain-derived neurotrophic factor, ciliary neurotrophic factor and others. Reactive astrocytes increase the expression of several of these, notably nerve growth factor, basic fibroblast growth factor, brain-derived neurotrophic factor and neuregulins, which can stimulate neurite outgrowth. Reactive astrocytes also overexpress neuropilin-1 and vascular endothelial growth factor, which act in concert to promote angiogenesis after cerebral ischemia. Hevin (SPARC-like protein 1), a synaptogenic protein released by astrocytes, forms a relay between a neurexin (pre-synaptic) and a neuroligin (post-synaptic); this relay is crucial for the synaptogenesis [89].

5.3. Modulating the function of astrocytes and heparin

5.3.1. Role of heparin in lysing the gliosis

Both unfractionated and low molecular weight heparins inhibit thrombin activation [90]. In addition, they have a fibrolytic (gliolytic) effect and can modulate astrocyte function.

Clinically, perineural application of condensed polytetrafluoroethylene-extractum cepae-heparin-allantoin gel during peripheral nerve surgery improves functional recovery [91]. The anti-fibrotic effects of heparin are well documented after flexor tendon surgery of the hand [92], in the resolution of intraperitoneal fibrosis [93, 94] and in improving various scar types [95].

Among other actions, heparan sulphate/heparin influences fibroblast growth factor responsible for cell proliferation, differentiation, signal transduction and angiogenesis [96, 97]. Heparin is known to inhibit fibroblast growth factor (FGF)-2-stimulated DNA synthesis as well as gene expression of FGF-2 and its receptor in AT2 pneumocytes [98]. Probably via syndecan-1, the presence of heparin at high concentrations reduces the activity of FGF-7, which is responsible for enhancement of keratinocytes migration and proliferation. Heparin enhances the action of FGF-1, which regulates the proliferation of fibroblasts, endothelial and epithelial cells, and influences angiogenesis via effect on the activity of endothelial cells. Heparin can enhance the stability of FGF-1 and might determine the formation of FGF1-FGFR (fibroblast growth factor receptor) active complex.

5.3.2. Possible role of heparin in modulating astrocyte function

Astrocyte stress response and trophic effects are mediated by the FGF family member, on which heparin exerts a profound influence [96, 99]. Fibroblast growth factor 1 (FGF1) has been shown to maintain the survival of neurons and induce neurite outgrowth [100]. Basic fibroblast growth factor (FGF-2) has been found to increase neuronal survival and neurite extension in foetal rat hippocampal neurons when bound to heparin substrates [101]. The length and sulphated position of heparin regulate FGF-2-dependent astrocytic transformation (stellation), native heparin significantly promoting FGF-2-dependent astrocytic transformation, whereas heparin hexasaccharide and 2-O-, 6-O- and N-desulphated heparins inhibit it [102]. Heparin affin regulatory peptide (HARP, pleiotrophin, heparin-binding growth-associated molecule) promotes neurite outgrowth and synaptic development. High levels of heparin affin regulatory peptide HARP mRNA and protein are induced in transformed astrocytes [103, 104]. Glypican-1 is a major high-affinity ligand of the Slit proteins, both of which are strongly upregulated in reactive astrocytes, suggesting their possible role in the inhibitory environment preventing axonal regeneration after injury. Heparins inhibit glypican-Slit interactions [105, 106].

5.4. Possible role of aspirin in modulating astrocyte function

Ciliary neurotrophic factor (CNTF) is a promyelinating trophic factor. Acetylsalicylic acid (aspirin) increases mRNA and protein expression of CNTF in primary mouse and human astrocytes in a dose- and time-dependent manner. Aspirin-induced astroglial CNTF is also functionally active; supernatants of aspirin-treated astrocytes of wild type, but not Cntf null, mice increase myelin-associated proteins in oligodendrocytes and protected oligodendrocytes from TNF-α insult [107].

5.5. Possible role of hyaluronic acid salts in modulating astrocyte function

The presence of high molecular weight hyaluronic acid (hyaluronic acid with limited degradation) after spinal cord injury decreases glial scarring. High molecular weight hyaluronic acid stabilised against degradation mitigates astrocyte activation in vitro and in vivo. Therefore, hyaluronic-acid-based hydrogel systems hold great potential for minimising undesired scarring as part of future repair strategies after spinal cord injury [108].

Advertisement

6. Combining peripheral nerve grafts with scaffolds: the scaffold as a drug release system

The defect within the spinal cord is too large to be bridged by nerve grafts alone; besides, the myelin sheath within them is inhibitory to axonal growth. The rationale for polymer implants is twofold, to replace a damaged area of the cord with just such a structural matrix [109] and to provide it with a synthetic scaffold, in which myelin is absent. Combining peripheral nerve grafts with scaffolds has gained more acceptance because of the importance of seeding the scaffolds with multiple nerve growth factor (NGF) spatial concentration gradients in order to promote axonal growth both within the scaffolds and the nerve grafts [22].

Biomaterial scaffolds in spinal cord injury have been reviewed by Madigan et al. [109] and Straley et al. [110]. Commonly used scaffolds include natural polymers (in vivo extracellular matrix polymers, polymers derived from blood, and polymers from marine life) and synthetic polymers (poly-hydroxy acid polymers and synthetic hydrogels). Examples for in vivo extracellular matrix polymers are collagen solutions and the glycosaminoglycan hyaluronic acid. Examples for polymers derived from blood are plasma-derived polymers, fibronectin and fibrin. Examples for polymers from marine life are agarose, alginate and chitosan. Synthetic polymers include poly-hydroxy acid polymers and synthetic hydrogels. Compared to natural polymers, they offer wider scope to design and control the characteristics of the material. Poly-hydroxy acid polymers are biodegradable materials based on polyesters of lactic and glycolic acid (PLA and PGA) and their co-polymer PLGA. Synthetic hydrogels are based on polyethylene glycol, a biodegradable synthetic polymer of ethylene oxide units. Poly(2-hydroxyethyl methacrylate) (pHEMA) compounds and pHEMA-co-methyl methacrylate (pHEMA-MMA) are used as spinal cord scaffolds.

Whatever macroengineering and microengineering procedures scaffolds are subjected to allow for axonal growth, this will not occur unless the scaffold is seeded with basal lamina and supplied with neurite outgrowth promoting factors and neurotrophic factors (Figure 3). These factors can be released from the scaffold material itself, from integrated micro- or nano-spheres or tubules of a different material, or by means of a scaffold’s capacity to support a genetically modified cell line in vivo [58, 109, 110].

Advertisement

7. Augmentation of peripheral nerve grafts with cellular transplants

Nearly half of the spinal cord injuries occur at the thoracolumbar junction (D12-L1), the site of the conus medullaris. We could potentially nerve graft the spinal cord (part of the CNS) directly to the cauda equina (part of the peripheral nervous system) without resorting to cellular transplants to restitute the neuronal and astrocytic components of the CNS. There is mounting evidence, however, that cellular transplants potentiate the effect of other factors and might even recruit endogenous neural precursor cells [62, 76].

7.1. Types of cellular transplants used to augment peripheral nerve grafts

In a meta-analysis reviewing cellular transplantation strategies in spinal cord injury, Tetzlaff et al. [111], have come to the following conclusions. Schwann cells are the most extensively studied cell type. They are reported both to remyelinate-injured spinal cord axons and to form a permissive substrate for their regeneration [112]. However, compared to neural precursors such as oligodendrocyte precursors or neural precursor/stem cells, they provoke a more robust astrocytic reaction, resulting in less effective integration into the host spinal cord [113]. Olfactory ensheathing cells demonstrate good integration into host spinal cord [114]. However, there is no robust evidence of improvement after their transplantation [115]. They also appear to require adjuvant treatment to increase efficacy (e.g., Schwann cells, Matrigel, rolipram, cAMP and neurotrophic factors) [116,117]. Neural stem/progenitor cells appear to integrate well into the host spinal cord with improved outcomes [118]. They differentiate mostly into astroglial cells, less so oligodendrocytes seen but rarely into neurons [119]. Suspicion has been raised as to their role in axonal regeneration [120, 121]. Fate-restricted neural and glial precursor have the potential to remyelinate injured axons [122]. More evidence is needed, however, to confirm this observation [123]. Bone marrow stromal cells have some bridging capacity in sharp transaction models [124]. However, their integration in the injured spinal cord is very limited. There is no convincing differentiation into neural cells despite claims to the contrary [125]. They are reported to stimulate neurite outgrowth over neural proteoglycans, myelin-associated glycoprotein and Nogo-A [126, 127].

7.2. Cellular transplants in combination strategy

Because of the previous controversies, the use of a combination strategy including Schwann cells has been recommended by Bunge [128]. The following combination strategy has been suggested [128]: Schwann cells, neuroprotective agents and growth factors administered in various ways, such as, olfactory ensheathing cell (OEC) implantation, chondroitinase addition or elevation of cyclic AMP. A targeted approach has been proposed by Kadoya et al. [43]. It includes the following: a peripheral conditioning lesion (bilateral sciatic nerve crush), mesenchymal stem cell transplantation mixed with neurotrophin-3 (NT-3) and creating a neurotrophic factor gradient by injection of lentivirus expressing neurotrophin-3 (NT-3) just proximal to the site of the lesion.

7.3. Number of cellular transplant injections

A third unresolved issue is the number of injections that the patient has to receive. Mackay-Sim et al. [129] have used a single intraoperative injection. Multiple injections have been recorded by other authors [130, 131].

7.4. Inducing mobilisation of neural precursor cells

Stem cell transplantation has the potential to recruit endogenous neural stem cells [132]. Neural stem cells exist in the mammalian developing and adult nervous system (mainly in the hippocamous and subventricular area). Multiple cell-intrinsic regulators coordinate neural stem cell maintenance, self-renewal and migration into injured areas. Essential intracellular regulators include the orphan nuclear receptor TLX, the high-mobility-group DNA binding protein Sox2, the basic helix-loop-helix transcription factor Hes, the tumour suppressor gene Pten, the membrane-associated protein Numb and its cytoplasmic homolog Numblike. Manipulating these factors among others [133135] by injecting them through indwelling catheters might induce mobilisation of neural stem cells to the injured spinal cord area.

Advertisement

8. Clinical application

8.1. Pre-operative assessment (neurological and radiographic evaluation)

Patients should be evaluated pre-operatively and at monthly intervals. Motor power and sensation should be evaluated using ASIA standards [28]. Confounding factors during motor power evaluation include fake muscle contractions produced by movements of the trunk and cocontractions between abdominal muscles and different muscle groups. Optional elements of ASIA neurologic impairment assessment should be included because the abdominal muscles and medial hamstrings are the first muscles to regain power.

Radiographic evaluation should include plain anteroposterior and lateral radiographs and pre-operative magnetic resonance imaging (MRI). The injury zone on the MRI is determined by the superior and inferior extents of the gliosis; nerve grafts have to be extended beyond the injury zone [136] (Figure 6).

Figure 6.

Cervical vertebral C5,6 fracture dislocation in a quadriplegic patient; the gliosis extends from the inferior border of cervical vertebra C4 to the superior border of cervical vertebra C6. The injury zone on the MRI is determined by the superior and inferior extents of the gliosis; nerve grafts have to be extended beyond the injury zone.

8.2. Timing of grafting

Spinal cord injury is considered chronic months to years after injury [6]. At this stage, the primary and secondary injuries have ceased. As the zone of gliosis may extend superiorly or inferiorly during the secondary injury phase, the patient should be operated upon at least 2 months after the injury, i.e., when it has become chronic.

Improvement is also independent of the time delay between the date of injury and the date of definitive surgery. This is supported by the observations made by Li and Raisman [137], who have noted that sprouts from cut corticospinal axons persist despite the presence of astrocytic scarring in long-term lesions of the adult rat spinal cord.

8.3. Operative technique, establishment of CSF circulation and establishment of a continuous drug delivery system (indwelling catheter implantation for post-operative drug and cell administration)

After exploring the injured cord through a posterior spinal laminectomy incision, sural nerves are side-grafted to the cord, especially on its ventral aspect [20, 138] (Figures 7(a and b) and 8). The cord graft construct invariably adheres to the dura preventing CSF circulation. The latter is important for nutrient, cell and growth factor transport [139, 140]. To prevent the cord adhering to the dura, it has become the author’s practice to wrap the cord graft construct with a silicone membrane (Figure 9). In fact, silicone chambers or tubes have been used as scaffolds for peripheral nerve regeneration [141143]. Interest to use them as scaffolds for cellular growth has been rekindled [144, 145]. They have been modified physically to increase porosity or coated to allow for growth of mesenchymal stem cells [146148] or even to allow neuron-like differentiation of mesenchymal stem cells [149]. They have been modified physically to increase porosity or chemically (with hyaluronic acid and hyaluronic acid--collagen conjugate) to allow for growth of neural cells [150, 151] or to inhibit glial tissue formation [152, 153]. To establish a continuous drug and cell delivery system, an indwelling percutaneous catheter is placed in the interstitium between the membrane and the dura; the dura is finally closed (Figure 9). Figure 10 is a schematic drawing of the hypothetical spinal cord-graft-scaffold-catheter construct.

Figure 7.

(a) Through a midline dorsal incision, a formal laminectomy is performed preserving the facet joints and pedicles. The dura is exposed. The dura is incised longitudinally and held with stay sutures exposing the cord lesion. The yellow arrows points to the defect in the cord. (b) Spinal cord lesion without defect but with a completely gliotic segment in a paraplegic patient suffering from a dorsolumbar fracture dislocation. The gliotic segment extends from the cord (white arrow) up to the cauda equine (yellow arrow).

Figure 8.

Sural nerve grafts having been side-grafted to the cord.

Figure 9.

To prevent the cord adhering to the dura, it has become the author’s practice to wrap the cord graft construct with a silicone membrane. To establish a continuous drug and cell delivery system, an indwelling catheter is placed in the interstitium between the membrane and the dura; the dura is finally closed.

Figure 10.

A schematic drawing of the hypothetical spinal cord-graft-scaffold-catheter construct: (I) end-to-end grafts; (II) side grafts; (III) synthetic scaffolds; (IV) cellular transplants; (V) modulated astrocytes; (VI) silicone membrane; (VII) dura mater; (VIII) indwelling catheter for post-operative delivery of neurolyzing agents (chondroitinase ABC, heparin), neurotrophic factors, neurite outgrowth promoting factors, injectable scaffolds and cellular transplants; (IX) skin.

8.4. Post-operative drug and cellular transplant administration through the catheter

The catheter can be used for post-operative administration of growth factors, neurolyzing agents, cellular transplants or even scaffolds. The author’s practice has been as follows. Starting from the fifth post-operative day calcium heparin (5000 IU) is injected every second day through the catheter. Chondroitinase ABC (5 IU, Sigma) is dissolved in 2 cc normal saline and injected on a weekly basis.

8.5. Catheter-related complications: keeping the indwelling catheter for 18 months or more around the spinal cord as a means of establishing a continuous spinal drug delivery system

Catheter-related complications include tension headache, meningitis, fibrous track formation, catheter slippage, difficult catheter insertion and catheter blockage. Fibrous track formation is noted by increased pressure on injection through the catheter, associated with increased serosanguinous discharge from the catheter skin exit site due to drug extrusion. Tension headache can be avoided by decreasing the volume of injection; meningitis and early catheter blockage and slippage are avoided by proper catheter care. Complications associated with fibrous track formation, such as difficult catheter insertion, late catheter blockage and slippage (during months 9–18) can only be avoided by inserting the catheter in the interstitium between the silicone membrane and the dura. In this way, the catheter need not be exchanged over 18 months.

8.6. Author’s clinical experience with heparin and chondroitinase ABC: its clinical safety and limitations

Delayed wound healing and sinus formation is related to repeated calheparin injection. Its incidence decreases when calheparin is administered every other day.

A vasovagal reaction occurs, when chondroitinase ABC is rapidly injected intrathecally. Its manifestations are cough, hypotension, general irritability and spinal cord irritability manifested by lower limb twitches. A vasovagal reaction does not occur, when the enzyme is injected extradurally or slowly intrathecally.

8.7. Results of surgery

In a clinical study [14], the right and left antero-lateral quadrant of the cord at T7-8 levels have been nerve grafted to homolateral L2-4 lumbar ventral roots. Eight months after surgery, voluntary contractions of bilateral adductors and of the left quadriceps have been observed.

Similar improvements have been observed in another study [15] after nerve side-grafting and augmentation by single-stage mesenchymal cell transplantation. Improvement has been hampered by cocontractions between abdominal muscles and different muscle groups. It has also been hampered by spasticity

In a not yet published study, the author has observed that repeated heparin, chondroitinase ABC and cellular transplant injection through an indwelling catheter placed in the interstitium between the membrane and the dura has led to the disappearance of cocontractions between abdominal muscles and different muscle groups. All patients have had pre-operative bouts of a moderate dull aching pain in the abdomen, back and both legs caused by adherence of the cord to the dura and the bony spinal canal. It has been completely resolved by inserting the silicone barrier membrane in the interstitium between the spinal cord and the dura.

Studies using cellular transplantation alone in spinal cord injuries have reported similar motor and sensory score improvement [154157]. In all these studies, spontaneous or treatment-induced anatomical neural plasticity as well as the adaptive reorganisation of the neural pathways occurring after injury and acting to restore some of the lost function have to be taken into consideration [4].

8.8. False positive results

On evaluating results of surgery after grafting the cord to the cauda equina in thoracolumbar lesions, it should be noted that false positive results could be obtained from intercostal nerves (peripheral nerves) regenerating into the cauda equina (peripheral nerves) via nerve grafts (Figure 11).

Figure 11.

In thoracolumbar lesions, false positive results could be obtained from intercostal nerves (peripheral nerves) at the cranial cord (white arrow) regenerating into to the caua equina (peripheral nerves) (yellow arrow) via nerve grafts.

Advertisement

9. Post-operative target organ derived trophic support

Target organ derived neurotrophic factors, the so-called neurotrophins (nerve growth factor(NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3(NT-3) and neurotrophin-4/5(NT-4/5)), are transported by retrograde axonal via an endosomal mechanism involving dyneins [158, 159]. Neurotrophins contribute a lot to axonal progression; in the injured spinal cord, this stimulus is lost. After grafting the spinal cord, an important question is whether this stimulus can be restituted by injecting neurotrophins into target organs post-operatively, or whether axonal progression into specific nerves can be restituted by injecting neurotrophins into the specific muscles supplied by them. Experimental evidence points to this [158, 159]. By the same token, it can be questioned whether other neurotrophic factors and neurolyzing agents can be similarly injected into target muscles. Both in vitro and in vivo local infusion of fibroblastic growth factors (FGFs) have been found to rescue motoneuron death induced by spinal cord injury [160]. However, evidence for retrograde axonal transport of heparin-binding growth factors is lacking [161].

Advertisement

10. Conclusions

We have outlined current experimental and clinical experience applying nerve side grafts to the injured spinal cord. Nerve side grafting increases the incidence of nerve regeneration by applying additional grafts extending from the side of the donor end of the cord to the side of the recipient end. A partially regenerated cord cannot be surgically cut and end grafted; nerve side grafting can enhance regeneration through it without incriminating already regained function. Nevertheless, side grafting will fail, unless the gliosis is counteracted or lysed by chondroitinase ABC, sialidase, anti-Nogo, Rho inhibitors and other factors. Side grafting will also fail unless neurons are stimulated to produce neurites. Modulating the function of astrocytes by heparin, aspirin and other factors is one method to stimulate the intrinsic properties of the neurons to produce neurites. Side grafting should be augmented by artificial scaffolds and cellular transplants. Clinically, to prevent the cord adhering to the dura and re-establish CSF circulation, it has become the author’s practice to wrap the cord graft construct with a silicone membrane. To establish a continuous drug and cell delivery system, an indwelling catheter is placed in the interstitium between the membrane and the dura; the dura is finally closed. Post-operative injection of paralysed muscles with neurotrophic factors stimulates neurite outgrowth by target-organ-derived neurotrophic support.

References

  1. 1. DeFelipe J, Jones EG: Cajal’s degeneration and regeneration of the nervous system (May RM, translator). New York: Oxford University Press, 1991.
  2. 2. Raisman G: A promising therapeutic approach to spinal cord repair. J R Soc Med 2003;96(6):259–261.
  3. 3. Samadikuchaksaraei A: An overview of tissue engineering approaches for management of spinal cord injuries. J Neuroeng Rehabil 2007;4:15.
  4. 4. Bradbury EJ, McMahon SB. Spinal cord repair strategies: why do they work? Nat Rev Neurosci 2006;7:644–653.
  5. 5. Reier PJ: Cellular transplantation strategies for spinal cord injury and translational neurobiology. Neurorx 2004 October;1(4):424–451.
  6. 6. Hyun JK, Kim HW. Clinical and experimental advances in regeneration of spinal cord injury. J Tissue Eng. 2010 Nov 2; Vol. 2010 : 1–20: Article ID 650857. doi: 10.4061/2010/650857. PubMed PMID: 21350645; PubMed Central PMCID: PMC3042682.
  7. 7. David S, Aguayo AJ: Axonal elongation into peripheral nervous system ´bridges´ after central nervous system injury in adult rats. Science 1981;214:931–933.
  8. 8. Tuszynski MH, Petersen DA, Ray J, Baird A, Nakahara Y, Gage FH: Fibroblasts genetically modified to produce nerve growth factor induce robust neuritic ingrowth after grafting to the spinal cord. Exp Neurol 1994;126:1–14.
  9. 9. Guenard V, Aebischer P, Bunge RP: The astrocyte inhibition of peripheral nerve regeneration is reversed by Schwann cells. Exp Neurol 1994;126:44–60.
  10. 10. Oudega M, Varon S, Hagg T: Regeneration of adult rat sensory axons into intraspinal nerve grafts: promoting effects of conditioning lesion and graft predegeneration. Exp Neurol 1994;129:194–206.
  11. 11. Liu S, Kadi K, Boisset N, Lacroix C, Said G, Tadie M: Reinnervation of denervated lumbar ventral roots and their target muscle by thoracic spinal motoneurons via an implanted nerve autograft in adult rats after spinal cord injury. J Neurosci Res 1999 Jun 1;56(5):506–517.
  12. 12. Liu S, Aghakhani N, Boisset N, Said G, Tadie M: Innervation of the caudal denervated ventral roots and their target muscles by the rostral spinal motoneurons after implanting a nerve autograft in spinal cord-injured adult marmosets. J Neurosurg 2001 Jan;94(1 Suppl):82–90.
  13. 13. Dam-Hieu P, Liu S, Choudhri T, Said G, Tadie M: Regeneration of primary sensory axons into the adult rat spinal cord via a peripheral nerve graft bridging the lumbar dorsal roots to the dorsal column. J Neurosci Res 2002 May 1;68(3):293–304.
  14. 14. Tadie M, Liu S, Robert R, Guiheneuc P, Pereon Y, Perrouin-Verbe B, Mathe JF: Partial return of motor function in paralyzed legs after surgical bypass of the lesion site by nerve autografts three years after spinal cord injury. J Neurotrauma 2002 Aug;19(8):909–916.
  15. 15. Amr SM, Gouda A, Koptan WT, Galal AA, Abdel-Fattah DS, Rashed LA, Atta HM, Abdel-Aziz MT: Bridging defects in chronic spinal cord injury using peripheral nerve grafts combined with a chitosan-laminin scaffold and enhancing regeneration through them by cotransplantation with bone-marrow derived mesenchymal stem cells; a series of 14 patients. J Spinal Cord Medicine 2014;37(1):54–71
  16. 16. Fawcett JW: Bridging spinal cord injuries. J Biol 2008;7:25.
  17. 17. Ray WZ: Mackinnon SE. Management of nerve gaps: autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol 2010 May ;223(1):77–85. doi:10.1016/j.expneurol.2009.03.031.
  18. 18. Frostick SP, Yin Q, Kemp G: Schwann cells, neurotrophic factors, and peripheral nerve regeneration. Microsurgery 1998;18:397–405.
  19. 19. Blottner D, Baumgarten HG: Neurotrophy and regeneration in vivo. Acta Anat 1994;150:235–245.
  20. 20. Amr SM, Moharram AN: Repair of brachial plexus lesions by end-to-side side-to-side grafting neurorrhaphy: experience based on 11 cases. Microsurgery 2005, 25(2):126–146.
  21. 21. Rosoff WJ, Urbach JS, Esrick MA, McAllister RG, Richards LJ, Goodhill GJ. A new chemotaxis assay shows the extreme sensitivity of axons to molecular gradients. Nat Neurosci 2004;7(6):678–682. Epub 2004 May 25. Erratum in: Nat Neurosci. 2004;7(7):785.
  22. 22. Tse TH, Chan BP, Chan CM, Lam J: Mathematical modeling of guided neurite extension in an engineered conduit with multiple concentration gradients of nerve growth factor (NGF). Ann Biomed Eng 2007;35(9):1561–1572. Epub 2007 May 23.
  23. 23. Ciofani G, Sergi PN, Carpaneto J, Micera S. A hybrid approach for the control of axonal outgrowth: preliminary simulation results. Med Biol Eng Comput 2011;49(2):163–170. doi: 10.1007/s11517-010-0687-x. Epub 2010 Oct 6. PubMed PMID: 20924708.
  24. 24. Ciofani G, Raffa V, Menciassi A, Micera S, Dario P: A drug delivery system based on alginate microspheres: mass-transport test and in vitro validation. Biomed Microdevices 2007;9:395–403.
  25. 25. Holmquist B, Kanje M, Kerns JM, Danielsen N. A mathematical model for regeneration rate and initial delay following surgical repair of peripheral nerves. J Neurosci Methods. 1993 Jun;Vol.48(1–2):27–33. PubMed PMID: 8377520.
  26. 26. Cover TM, Thomas JA: Elements of Information Theory, 2nd ed. Hoboken, New Jersey: John Wiley & Sons, Inc., 2006.
  27. 27. Effenberger F. A Primer on Information Theory, with Applications to Neuroscience. In: Rakocevic G., Djukic T., Filipovic N., Milutinović V. (editors) Computational Medicine in Data Mining and Modeling. New York: Springer 2013.
  28. 28. Standards for Neurological Classification of Spinal Injury. American Spinal Injury Association (ASIA) Illinois: Chicago, 1996.
  29. 29. Harel NY, Strittmatter SM: Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? Nat Rev Neurosci 2006;7:603–616.
  30. 30. Côté MP, Amin AA, Tom VJ, Houle JD: Peripheral nerve grafts support regeneration after spinal cord injury. Neurotherapeutics 2011;8(2):294–303.
  31. 31. Hill CE, Brodak DM, Bartlett Bunge M: Dissociated predegenerated peripheralnerve transplants for spinal cord injury repair: a comprehensive assessment of their effects on regeneration and functional recovery compared to Schwann cell transplants. J Neurotrauma 2012;29(12):2226–2243. doi: 10.1089/neu.2012.2377. PubMed PMID: 22655857; PubMed Central PMCID: PMC3472680.
  32. 32. Bray GM, Villegas-Perez MP, Vidal-Sanz M, Aguayo AJ: The use of peripheral nerve grafts to enhance neuronal survival, promote growth and permit terminal reconnections in the central nervous system of adult rats. J Exp Biol 1987;132:5–19.
  33. 33. Bunge MB: Bridging areas of injury in the spinal cord. Neuroscientist 2001;7:325–339.
  34. 34. Guzen FP, de Almeida Leme RJ, de Andrade MS, de Luca BA, Chadi G: Glial cell line-derived neurotrophic factor added to a sciatic nerve fragment grafted in a spinal cord gap ameliorates motor impairments in rats and increases local axonal growth. Restor Neurol Neurosci 2009;27(1):1–16.
  35. 35. Cheng H, Cao Y, Olson L: Spinal cord repair in adult paraplegic rats partial restoration of hind limb function. Science 1996;273:510–513.
  36. 36. Lee YS, Hsiao I, Lin VW: Peripheral nerve grafts and aFGF restore partial hindlimb function in adult paraplegic rats. J Neurotrauma 2002;19:1203–1216.
  37. 37. Campos L, Meng Z, Hu G, Chiu DT, Ambron RT, Martin JH: Engineering novel spinal circuits to promote recovery after spinal injury. J Neurosci 2004;24:2090–2101.
  38. 38. Gauthier P, Rega P, Lammari-Barreault N, Polentes J: Functional reconnections established by central respiratory neurons regenerating axons into a nerve graft bridging the respiratory centers to the cervical spinal cord. J Neurosci Res 2002;70:65–81.
  39. 39. Decherchi P, Lammari-Barreault N, Gauthier P: Regeneration of respiratory pathways within spinal peripheral nerve grafts. Exp Neurol 1996;137:1–14.
  40. 40. Aguayo AJ, Bray GM, Rasminsky MM, Zwimpfer T, Carter D, Vidal-Sanz M: Synaptic connections made by axons regenerating in the central nervous system of adult mammals. J Exp Biol 1990;153:199–224.
  41. 41. Plant GW, Bates ML, Bunge MB: Inhibitory proteoglycan immunoreactivity is higher at the caudal than the rostral Schwann cell graft-transected spinal cord interface. Mol Cell Neurosci 2001;17:471–487.
  42. 42. Thuret S, Moon LDF, Gage FH: Therapeutic interventions after spinal cord injury. Nat Rev Neurosci 2006;7:628–643.
  43. 43. Kadoya K, Tsukada S, Lu P, Coppola G, Geschwind D, Filbin MT, Blesch A,Tuszynski MH: Combined intrinsic and extrinsic neuronal mechanisms facilitate bridging axonal regeneration one year after spinal cord injury. Neuron. 2009;64(2):165–172. doi: 10.1016/j.neuron.2009.09.016. PubMed PMID: 19874785; PubMedCentral PMCID: PMC2773653.
  44. 44. Neumann S, Bradke F, Tessier-Lavigne M, Basbaum AI: Regeneration of sensory axons within the injured spinal cord induced by intraganglionic cAMP elevation. Neuron. 2002;34:885–893 [PubMed].
  45. 45. Qiu J, Cai D, Dai H, McAtee M, Hoffman PN, Bregman BS, Filbin MT: Spinal axon regeneration induced by elevation of cyclic AMP. Neuron 2002;34:895–903 [PubMed].
  46. 46. Ferguson TA, Son YJ: Extrinsic and intrinsic determinants of nerve regeneration. J Tissue Eng 2011;2(1):2041731411418392. doi: 10.1177/2041731411418392. Epub 2011 Sep 13. PubMed PMID: 22292105; PubMed Central PMCID: PMC3251917.
  47. 47. 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 2004;24(29):6531–6539. PubMed PMID: 15269264.
  48. 48. Blits B, Carlstedt TP, Ruitenberg MJ, de Winter F, Hermens WT, Dijkhuizen PA, Claasens JW, Eggers R, Sluis R van der, Tenenbaum L, Boer GJ, Verhaagen J: Rescue and sprouting of motoneurons following ventral root avulsion and reimplantation combined with intraspinal adeno-associated viral vector-mediated expression of glial cell line-derived neurotrophic factor or brain-derived neurotrophic factor. Exp Neurol 2004;189(2):303–316.
  49. 49. Yiu G, He Z: Glial inhibition of CNS axon regeneration. Nat Rev Neurosci 2006;7:617–627.
  50. 50. McCreedy DA, Sakiyama-Elbert SE: Combination therapies in the CNS: engineering the environment. Neurosci Lett 2012;519(2):115–121. doi: 10.1016/j.neulet.2012.02.025. Epub 2012 Feb 17. Review. PMID: 22343313.
  51. 51. Bradbury EJ, Carter LM: Manipulating the glial scar: chondroitinase ABC as a therapy for spinal cord injury. Brain Res Bull 2011;84:306316 [PubMed: 20620201].
  52. 52. Galtrey CM, Fawcett JW: The role of chondroitin sulfate proteoglycans in regeneration and plasticity in the central nervous system. Brain Res Rev 2007;54:118 [PubMed: 17222456].
  53. 53. Yamagata T, Saito H, Habuchi O, Suzuki S: Purification and properties of bacterial chondroitinases and chondrosulfatases. J Biol Chem 1968;243:15231535 [PubMed: 5647268].
  54. 54. 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 [PubMed: 11948352].
  55. 55. 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(28):7405–7415.
  56. 56. Tom VJ, Sandrow-Feinberg HR, Miller K, Santi L, Connors T, Lemay MA, Houle JD: Combining peripheral nerve grafts and chondroitinase promotes functional axonal regeneration in the chronically injured spinal cord. J Neurosci 2009;29:14881–14890 [PMC free article] [PubMed].
  57. 57. Hwang DH, Kim HM, Kang YM, Joo IS, Cho CS, Yoon BW, Kim SU, Kim BG. Combination of multifaceted strategies to maximize the therapeutic benefits of neural stem cell transplantation for spinal cord repair. Cell Transplant. 2011;20(9):1361–1379. doi: 10.3727/096368910X557155. Epub 2011 Mar 7. PubMed PMID: 21396156.
  58. 58. Kumar P, Choonara YE, Modi G, Naidoo D, Pillay V: Multifunctional therapeutic delivery strategies for effective neuro-regeneration following traumatic spinal cord injury. Curr Pharm Des 2015;21(12):1517–1528 [PubMed PMID: 25594407].
  59. 59. Jain A, Kim YT, McKeon RJ, Bellamkonda RV: In situ gelling hydrogels for conformal repair of spinal cord defects, and local delivery of BDNF after spinal cord injury. Biomaterials 2006;27:497504 [PubMed: 16099038].
  60. 60. Kreider BQ, Messing A, Doan H, Kim SU, Lisak RP, Pleasure DE. Enrichment of Schwann cell cultures from neonatal rat sciatic nerve by differential adhesion. Brain Res 1981;207:433444 [PubMed: 7008901].
  61. 61. Fouad K, Schnell L, Bunge MB, Schwab ME, Liebscher T, Pearse DD. Combining Schwann cell bridges and olfactory-ensheathing glia grafts with chondroitinase promotes locomotor recovery after complete transection of the spinal cord. J Neurosci 2005;25:1169–1178 [PubMed].
  62. 62. Wilems TS, Pardieck J, Iyer N, Sakiyama-Elbert SE: Combination therapy of stem cell derived neural progenitors and drug delivery of anti-inhibitory molecules for spinal cord injury. Acta Biomater 2015. pii: S1742–7061(15)30112–4. doi: 10.1016/j.actbio.2015.09.018. [Epub ahead of print]
  63. 63. Lee H, McKeon RJ, Bellamkonda RV. Sustained delivery of thermostabilized chABC enhances axonal sprouting and functional recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2010 Feb 23;107(8):3340–3345. doi: 10.1073/pnas.0905437106. Epub 2009 Nov 2. PubMed PMID: 19884507; PubMed Central PMCID: PMC2840440.
  64. 64. Tom VJ, Houlé JD: Intraspinal microinjection of chondroitinase ABC following injury promotes axonal regeneration out of a peripheral nerve graft bridge. Exp Neurol 2008;211(1):315–319.
  65. 65. Wilems TS, Sakiyama-Elbert SE: Sustained dual drug delivery of anti-inhibitory molecules for treatment of spinal cord injury. J Control Release 2015;213:103–111. doi: 10.1016/j.jconrel.2015.06.031 [Epub ahead of print] [PubMed PMID: 26122130].
  66. 66. Hyatt AJ, Wang D, Kwok JC, Fawcett JW, Martin KR: Controlled release of chondroitinase ABC from fibrin gel reduces the level of inhibitory glycosaminoglycan chains in lesioned spinal cord. J Control Release 2010;147(1):24–29. doi: 10.1016/j.jconrel.2010.06.026.
  67. 67. Hunanyan AS, García-Alías G, Alessi V, Levine JM, Fawcett JW, Mendell LM, Arvanian VL: Role of chondroitin sulfate proteoglycans in axonal conduction in Mammalian spinal cord. J Neurosci 2010;30(23):7761–7769. doi: 10.1523/JNEUROSCI.4659-09.2010.
  68. 68. Cheng CH, Lin CT, Lee MJ, Tsai MJ, Huang WH, Huang MC, Lin YL, Chen CJ, Huang WC, Cheng H: Local delivery of high-dose chondroitinase ABC in the sub-acute stage promotes axonal outgrowth and functional recovery after complete spinal cord transection. PLoS One. 2015;10(9):e0138705. doi:10.1371/journal.pone.0138705. eCollection 2015.
  69. 69. Shields LB, Zhang YP, Burke DA, Gray R, Shields CB: Benefit of chondroitinase ABC on sensory axon regeneration in a laceration model of spinal cord injury in the rat. Surg Neurol 2008;69(6):568–577; discussion 577. doi: 10.1016/j.surneu.2008.02.009.
  70. 70. Iseda T, Okuda T, Kane-Goldsmith N, Mathew M, Ahmed S, Chang YW, Young W, Grumet M: Single, high-dose intraspinal injection of chondroitinase reduces glycosaminoglycans in injured spinal cord and promotes corticospinal axonal regrowth after hemisection but not contusion. J Neurotrauma 2008;25(4):334–349. doi: 10.1089/neu.2007.0289.
  71. 71. Caggiano AO, Zimber MP, Ganguly A, Blight AR, Gruskin EA: Chondroitinase ABCI improves locomotion and bladder function following contusion injury of the rat spinal cord. J Neurotrauma 2005;22(2):226–239.
  72. 72. Mondello SE, Jefferson SC, Tester NJ, Howland DR: Impact of treatment duration and lesion size on effectiveness of chondroitinase treatment post-SCI. Exp Neurol 2015;267:64–77. doi: 10.1016/j.expneurol.2015.02.028. Epub 2015 Feb 26.
  73. 73. Ma Y, Liu M, Li Y: Secretion of bacterial chondroitinase ABC from bone marrow stromal cells by glycosylation site mutation: a promising approach for axon regeneration. Med Hypotheses 2011;77(5):914–916. doi: 10.1016/j.mehy.2011.08.010. Epub 2011 Aug 31.
  74. 74. Kumar P, Choonara YE, Modi G, Naidoo D, Pillay V(1): Multifunctional therapeutic delivery strategies for effective neuro-regeneration following traumatic spinal cord injury. Curr Pharm Des 2015;21(12):1517–1528.
  75. 75. 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(35):8066–8076.
  76. 76. Sarveazad A, Bakhtiari M, Babahajian A, Janzade A, Fallah A, Moradi F, Soleimani M, Younesi M, Goudarzi F, Mohammad Taghi Joghataei: Comparison of human adipose-derived stem cells and chondroitinase ABC transplantation on locomotor recovery in the contusion model of spinal cord injury in rats. Iran J Basic Med Sci 2014;17(9):685–693. PubMed PMID: 25691946; PubMed Central PMCID:PMC4322153.
  77. 77. Mountney A, Zahner MR, Sturgill ER, Riley CJ, Aston JW, Oudega M, Schramm LP, Hurtado A, Schnaar RL: Sialidase, chondroitinase ABC, and combination therapy after spinal cord contusion injury. J Neurotrauma 2013;30(3):181–190. doi: 10.1089/neu.2012.2353
  78. 78. Lang BT, Cregg JM, DePaul MA, Tran AP, Xu K, Dyck SM, Madalena KM, Brown BP, Weng YL, Li S, Karimi-Abdolrezaee S, Busch SA, Shen Y, Silver J: Modulation of the proteoglycan receptor PTPσ promotes recovery after spinal cord injury. Nature 2015;518(7539):404–408. doi: 10.1038/nature13974. Epub 2014 Dec 3. PubMed PMID: 25470046; PubMed Central PMCID: PMC4336236.
  79. 79. Silver L, Michael JV, Goldfinger LE, Gallo G: Activation of PI3K and R-Ras signaling promotes the extension of sensory axons on inhibitory chondroitin sulfate proteoglycans. Dev Neurobiol 2014;74(9):918–933. doi: 10.1002/dneu.22174. Epub 2014 Mar 27.
  80. 80. Tetzlaff W, Alexander SW, Miller FD, Bisby MA: Response of facial and rubrospinal neurons to axotomy: changes in mRNA expression for cytoskeletal proteins and GAP-43. J Neurosci 1991;11:2528–2544 [PubMed].
  81. 81. Sengottuvel V, Leibinger M, Pfreimer M, Andreadaki A, Fischer D: Taxol facilitates axon regeneration in the mature CNS. J Neurosci 2011;31:2688–2699 [PubMed].
  82. 82. Bates CA, Becker CG, Miotke JA, Meyer RL: Expression of polysialylated NCAM but not L1 or N-cadherin by regenerating adult mouse optic fibers in vitro. Exp Neurol 1999;155:128–139 [PubMed].
  83. 83. Chaisuksunt V, Zhang Y, Anderson PN, et al: Axonal regeneration from CNS neurons in the cerebellum and brainstem of adult rats: correlation with the patterns of expression and distribution of messenger RNAs for L1, CHL1, c-jun and growth-associated protein-43. Neuroscience 2000;100:87–108 [PubMed].
  84. 84. Chen J, Wu J, Apostolova I, et al: Adeno-associated virus-mediated L1 expression promotes functional recovery after spinal cord injury. Brain 2007;130:954–969 [PubMed].
  85. 85. Roonprapunt C, Huang W, Grill R, et al: Soluble cell adhesion molecule L1-Fc promotes locomotor recovery in rats after spinal cord injury. J Neurotrauma 2003;20:871–882 [PubMed].
  86. 86. Chen Y and Swanson RA: Astrocytes and brain injury. Rev Article J Cereb Blood Flow Metab 2003;23(2):137–149.
  87. 87. Mohn TC, Koob AO: Adult astrogenesis and the etiology of cortical neurodegeneration. J Exp Neurosci 2015:9(s) 25–34. doi:10.4137/JEn.s25520.
  88. 88. Hering TM, Beller JA, Calulot CM, Centers A, Snow DM: Proteoglycans of reactive rat cortical astrocyte cultures: abundance of N-unsubstituted glucosamine-enriched heparan sulfate. Matrix Biol 2015;41:8–18. doi: 10.1016/j.matbio.2014.11.006.
  89. 89. Yates D: Synaptogenesis. Asynaptic bridge. Nat Rev Neurosci 2016;17,135. doi:10.1038/nrn.2016.12 Published online 11 February 2016.
  90. 90. Harter K, Levine M, Henderson SO: Anticoagulation drug therapy: a review. West J Emerg Med 2015;16(1):11–17. doi: 10.5811/westjem.2014.12.22933. Epub 2015 Jan 12. Review. PubMed PMID: 25671002; PubMed Central PMCID: PMC4307693.
  91. 91. Kahraman A, Kahveci R: Evaluating the effect of polytetrafluoroethylene and extractum cepae-heparin-allantoin gel in peripheral nerve injuries in a rat model. Plast Surg (Oakv) 2015 Spring;23(1):9–14. PubMed PMID: 25821766; PubMed Central PMCID: PMC4364148.
  92. 92. Akbari H, Rahimi AA, Ghavami Y, Mousavi SJ, Fatemi MJ. Effect of heparin on post-operative adhesion in flexor tendon surgery of the hand. J Hand Microsurg 2015;7(2):244–249. Epub 2015 Aug 26. PubMed PMID: 26578825; PubMed Central PMCID: PMC4642480.
  93. 93. Aydoseli A, Tahta A, Aras Y, Sabancı A, Keskin M, Balik E, Onder S, Sencer A, Izgi N: Use of antifibrotics to prevent ventriculoperitoneal shunt complications due to intra-abdominal fibrosis: experimental study in a rat model. J Neurol Surg A Cent Eur Neurosurg 2015;76(3):219–223. doi: 10.1055/s-0034-1389369. Epub 2015 Mar 26. PubMed PMID: 25811104.
  94. 94. Alonso Jde M, Rodrigues KA, Yamada AL, Watanabe MJ, Alves AL, Rodrigues CA, Hussni CA: Peritoneal reactivity evaluation in horses subjected to experimental small colon enterotomy and treated with subcutaneous heparin. Vet Med Int 2014;2014:385392. doi: 10.1155/2014/385392. Epub 2014 Nov 11. PubMed PMID: 25436172; PubMed Central PMCID: PMC4243600.
  95. 95. Ho WS, Ying SY, Chan PC, Chan HH: Use of onion extract, heparin, allantoin gel in prevention of scarring in chinese patients having laser removal of tattoos: a prospective randomized controlled trial. Dermatol Surg 2006;32(7):891–896. PubMed PMID: 16875470.
  96. 96. Billings PC, Pacifici M: Interactions of signaling proteins, growth factors and other proteins with heparan sulfate: mechanisms and mysteries. Connect Tissue Res 2015;56(4):272–280. doi: 10.3109/03008207.2015.1045066.
  97. 97. Olczyk P, Mencner Ł, Komosinska-Vassev K: Diverse roles of heparan sulfate and heparin in wound repair. Biomed Res Int 2015;2015:549417. doi: 10.1155/2015/549417. Epub 2015 Jul 7. Review. PubMed PMID: 26236728; PubMed Central PMCID: PMC4508384.
  98. 98. Newman DR, Li CM, Simmons R, Khosla J, Sannes PL: Heparin affects signaling pathways stimulated by fibroblast growth factor-1 and -2 in type II cells. Am J Physiol Lung Cell Mol Physiol 2004;287(1):L191–L200. Epub 2004 Feb 13. PubMed PMID: 14966081.
  99. 99. Santos-Silva A, Fairless R, Frame MC, Montague P, Smith GM, Toft A, Riddell JS, Barnett SC: FGF/heparin differentially regulates Schwann cell and olfactory ensheathing cell interactions with astrocytes: a role in astrocytosis. J Neurosci 2007;27(27):7154–7167.
  100. 100. Yi-Chao Hsu, Su-Liang Chen, Dan-Yen Wang: Ing-Ming Chiu stem cell-based therapy in neural repair. Biomed J 2013;36:98–105.
  101. 101. Walicke P, Cowan WM, Ueno N, Baird A, Guillemin R: Fibroblast growth factor promotes survival of dissociated hippocampal neurons and enhances neurite extension. Proc Natl Acad Sci U S A 1986;83(9):3012–3016.
  102. 102. Nagayasu T, Miyata S, Hayashi N, Takano R, Kariya Y, Kamei K: Heparin structures in FGF-2-dependent morphological transformation of astrocytes. J Biomed Mater Res A 2005;74(3):374–380. PubMed PMID: 15973728.
  103. 103. Poulsen FR, Lagord C, Courty J, Pedersen EB, Barritault D, Finsen B: Increased synthesis of heparin affin regulatory peptide in the perforant path lesioned mouse hippocampal formation. Exp Brain Res 2000;135(3):319–330. PubMed PMID: 11146810.
  104. 104. Wanaka A, Carroll SL, Milbrandt J: Developmentally regulated expression of pleiotrophin, a novel heparin binding growth factor, in the nervous system of the rat. Brain Res Dev Brain Res 1993;72(1):133–144. PubMed PMID: 8453763.
  105. 105. Hagino S, Iseki K, Mori T, Zhang Y, Sakai N, Yokoya S, Hikake T, Kikuchi S, Wanaka A: Expression pattern of glypican-1 mRNA after brain injury in mice. Neurosci Lett 2003;349(1):29–32. PubMed PMID: 12946579.
  106. 106. Lau E, Margolis RU: Inhibitors of slit protein interactions with the heparin sulphate proteoglycan glypican-1: potential agents for the treatment of spinal cord injury. Clin Exp Pharmacol Physiol 2010;37(4):417–421. doi: 10.1111/j.1440-1681.2009.05318.x.
  107. 107. Modi KK, Sendtner M, Pahan K: Up-regulation of ciliary neurotrophic factor in astrocytes by aspirin: implications for remyelination in multiple sclerosis. J Biol Chem 2013;288(25):18533–18545. doi: 10.1074/jbc.M112.447268.
  108. 108. Khaing ZZ, Milman BD, Vanscoy JE, Seidlits SK, Grill RJ, Schmidt CE: High molecular weight hyaluronic acid limits astrocyte activation and scar formation after spinal cord injury. J Neural Eng 2011;8(4):046033. doi: 10.1088/1741-2560/8/4/046033.
  109. 109. Madigan NN, McMahon S, O'Brien T, Yaszemski MJ, Windebank AJ: Current tissue engineering and novel therapeutic approaches to axonal regeneration following spinal cord injury using polymer scaffolds. Respir Physiol Neurobiol 2009;169(2):183–199. doi: 10.1016/j.resp.2009.08.015. Epub 2009 Sep 6. Review. PubMed PMID: 19737633; PubMed Central PMCID: PMC2981799.
  110. 110. Straley KS, Foo CW, Heilshorn SC: Biomaterial design strategies for the treatment of spinal cord injuries. J Neurotrauma 2010;27(1):1–19. doi: 10.1089/neu.2009.0948. Review. PubMed PMID: 19698073; PubMed Central PMCID: PMC2924783.
  111. 111. Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS, Plemel JR, Plunet WT, Tsai EC, Baptiste D, Smithson LJ, Kawaja MD, Fehlings MG, Kwon BK: A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma 2011;28(8):1611–1682. Epub 2010 Apr 20. Review.
  112. 112. Duncan ID, Milward EA: Glial cell transplants: experimental therapies of myelin diseases. Brain Pathol 1995;5:301–310 [PubMed].
  113. 113. Santos-Silva A, Fairless R, Frame MC, Montague P, Smith GM, Toft A, Riddell JS, Barnett SC: FGF/heparin differentially regulates Schwann cell and olfactory ensheathing cell interactions with astrocytes: a role in astrocytosis. J Neurosci 2007;27(27):7154–7167.
  114. 114. Ramon-Cueto A, Plant GW, Avila J, Bunge MB: Long-distance axonal regeneration in the transected adult rat spinal cord is promoted by olfactory ensheathing glia transplants. J Neurosci 1998;18:3803–3815 [PubMed].
  115. 115. Lu P, Yang H, Culbertson M, Graham L, Roskams AJ, Tuszynski MH: Olfactory ensheathing cells do not exhibit unique migratory or axonal growth-promoting properties after spinal cord injury. J Neurosci 2006;26:11120–11130 [PubMed].
  116. 116. Pearse DD, Sanchez AR, Pereira FC, Andrade CM, Puzis R, Pressman Y, Golden K, Kitay BM, Blits B, Wood PM, Bunge MB: Transplantation of Schwann cells and/or olfactory ensheathing glia into the contused spinal cord: survival, migration, axon association, and functional recovery. Glia 2007;55:976–1000 [PubMed].
  117. 117. Ruitenberg MJ, Plant GW, Hamers FP, Wortel J, Blits B, Dijkhuizen PA, Gispen WH, Boer GJ, Verhaagen J: Ex vivo adenoviral vector-mediated neurotrophin gene transfer to olfactory ensheathing glia: effects on rubrospinal tract regeneration, lesion size, and functional recovery after implantation in the injured rat spinal cord. J Neurosci 2003;23:7045–7058 [PubMed].
  118. 118. Alexanian AR, Crowe MJ, Kurpad SN: Efficient differentiation and integration of lineage-restricted neural precursors in the traumatically injured adult cat spinal cord. J Neurosci Methods 2006;150:41–46 [PubMed].
  119. 119. Cao QL, Zhang YP, Howard RM, Walters WM, Tsoulfas P, Whittemore SR: Pluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord are restricted to a glial lineage. Exp Neurol 2001;167:48–58 [PubMed].
  120. 120. Macias MY, Syring MB, Pizzi MA, Crowe MJ, Alexanian AR, Kurpad SN: Pain with no gain: allodynia following neural stem cell transplantation in spinal cord injury. Exp Neurol 2006;201:335–348 [PubMed].
  121. 121. Jin Y, Bouyer J, Shumsky JS, Haas C, Fischer I: Transplantation of neural progenitor cells in chronic spinal cord injury. Neuroscience 2016;320:69–82. doi: 10.1016/j.neuroscience.2016.01.066. Epub 2016 Feb 4. PubMedPMID: 26852702.
  122. 122. Bambakidis NC, Miller RH: Transplantation of oligodendrocyte precursors and sonic hedgehog results in improved function and white matter sparing in the spinal cords of adult rats after contusion. Spine J 2004;4:16–26 [PubMed].
  123. 123. Cao QL, Howard RM, Dennison JB, Whittemore SR. Differentiation of engrafted neuronal-restricted precursor cells is inhibited in the traumatically injured spinal cord. Exp Neurol 2002;177:349–359 [PubMed].
  124. 124. Ankeny DP, McTigue DM, Jakeman LB: Bone marrow transplants provide tissue protection and directional guidance for axons after contusive spinal cord injury in rats. Exp Neurol 2004;190:17–31 [PubMed].
  125. 125. Yano S, Kuroda S, Lee JB, Shichinohe H, Seki T, Ikeda J, Nishimura G, Hida K, Tamura M, Iwasaki Y: In vivo fluorescence tracking of bone marrow stromal cells transplanted into a pneumatic injury model of rat spinal cord. J Neurotrauma 2005;22:907–918 [PubMed].
  126. 126. Wright KT, Masri WE, Osman A, Roberts S, Trivedi J, Ashton BA, Johnson WE: The cell culture expansion of bone marrow stromal cells from humans with spinal cord injury: implications for future cell transplantation therapy. Spinal Cord 2008;46(12):811–817.
  127. 127. Wright KT, El Masri W, Osman A, Roberts S, Chamberlain G, Ashton BA, Johnson WE: Bone marrow stromal cells stimulate neurite outgrowth over neural proteoglycans (CSPG), myelin associated glycoprotein and Nogo-A. Biochem Biophys Res Commun 2007;354(2):559–566.
  128. 128. Bunge MB: Novel combination strategies to repair the injured mammalian spinal cord. J Spinal Cord Med 2008;31:262–269.
  129. 129. Mackay-Sim A, Feron F, Cochrane J, Bassingthwaighte L, Bayliss C, Davies W, Fronek P, Gray C, Kerr G, Licina P, Nowitzke A, Perry C, Silburn PAS, Urquhart S, Geraghty T: Autologous olfactory ensheathing cell transplantation in human paraplegia: a 3-year clinical trial. Brain 2008;131:2376–2386.
  130. 130. Li H, Wen Y, Luo Y, Lan X, Wang D, Sun Z, Hu L: [Transplantation of bone marrow mesenchymal stem cells into spinal cord injury: a comparison of delivery different times] [Article in Chinese] Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2010;24(2):180–184.
  131. 131. Ichim TE, Solano F, Lara F, Paris E, Ugalde F, Rodriguez JP, Minev B, Bogin V, Ramos F, Woods EJ, Murphy MP, Patel AN, Harman RJ, Riordan NH: Feasibility of combination allogeneic stem celltherapy for spinal cord injury: a case report. Int Archives Med 2010;3:30.
  132. 132. Sullivan R, Duncan K, Dailey T, Kaneko Y, Tajiri N, Borlongan CV: A possible new focus for stroke treatment—migrating stem cells. Expert Opin Biol Ther 2015;15(7):949–958. doi: 10.1517/14712598.2015.1043264.
  133. 133. Klingener M, Chavali M, Aguirre A: N-cadherin promotes recruitment and migration of neural progenitor cells from the SVZ stem cell niche into demyelinated lesions. Int J Dev Neurosci 2015;47(Pt A):107. doi: 10.1016/j.ijdevneu.2015.04.292. PubMed PMID: 26531615.
  134. 134. Kim HJ, Shaker MR, Cho B, Cho HM, Kim H, Kim JY, Sun W: Dynamin-related protein 1 controls the migration and neuronal differentiation of subventricular zone-derived neural progenitor cells. Sci Rep 2015;5:15962. doi: 10.1038/srep15962.
  135. 135. Chen Q, Zhang M, Li Y, Xu D, Wang Y, Song A, Zhu B, Huang Y, Zheng JC: CXCR7 mediates neural progenitor cells migration to CXCL12 independent of CXCR4. Stem Cells 2015;33(8):2574–2585. doi: 10.1002/stem.2022. Epub 2015 May 13. PubMed PMID: 25833331.
  136. 136. Potter K, Saifuddin A: MRI of chronic spinal cord injury. Br J Radiol 2003;76:347–352.
  137. 137. 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.
  138. 138. Amr SM, Essam AM, Abdel-Meguid AM, Kholeif AM, Moharram AN, El-Sadek RE: Direct cord implantation in brachial plexus avulsions: revised technique using a single stage combined anterior (first) posterior (second) approach and end-to-side side-to-side grafting neurorrhaphy. J Brachial Plex Peripher Nerve Inj 2009;4:8.
  139. 139. Nakagomi T, Molnár Z, Nakano-Doi A, Taguchi A, Saino O, Kubo S, Clausen M, Yoshikawa H, Nakagomi N, Matsuyama T: Ischemia-induced neural stem/progenitor cells in the pia mater following cortical infarction. Stem Cells Dev 2011 Dec;20(12):2037–2051.
  140. 140. Johanson C, Stopa E, Baird A, Sharma H: Traumatic brain injury and recovery mechanisms: peptide modulation of periventricular neurogenic regions by the choroid plexus-CSF nexus. J Neural Transm 2011;118(1):115–133. Epub 2010 Oct 10.
  141. 141. Francel PC, Francel TJ, Mackinnon SE, Hertl C: Enhancing nerve regeneration across a silicone tube conduit by using interposed short-segment nerve grafts. J Neurosurg 1997;87(6):887–892. PubMed PMID: 9384400.
  142. 142. Spector JG, Lee P, Derby A, Roufa DG: Comparison of rabbit facial nerve regeneration in nerve growth factor-containing silicone tubes to that in autologous neural grafts. Ann Otol Rhinol Laryngol 1995;104(11):875–885.PubMed PMID: 8534028.
  143. 143. Le Beau JM, Powell HC, Ellisman MH: Node of Ranvier formation along fibres regenerating through silicone tube implants: a freeze-fracture and thin-section electron microscopic study. J Neurocytol 1987;16(3):347–358. PubMed PMID: 3612184.
  144. 144. Peláez RJ, Afonso CN, Vega F, Recio-Sánchez G, Torres-Costa V, Manso-Silván M, García-Ruiz JP, Martín-Palma RJ: Laser fabrication of porous silicon-based platforms for cell culturing. J Biomed Mater Res B Appl Biomater 2013;101(8):1463–1468. doi: 10.1002/jbm.b.32966.
  145. 145. Punzón-Quijorna E, Sánchez-Vaquero V, Muñoz-Noval A, Pérez-Roldán MJ, Martín-Palma RJ, Rossi F, Climent-Font A, Manso-Silván M, Ruiz JP, Torres-Costa V: Nanostructured porous silicon micropatterns as a tool forsubstrate-conditioned cell research. Nanoscale Res Lett 2012;7(1):396. PubMed PMID: 22799489; PubMed Central PMCID: PMC3458952.
  146. 146. Collart-Dutilleul PY, Panayotov I, Secret E, Cunin F, Gergely C, Cuisinier F, Martin M: Initial stem cell adhesion on porous silicon surface: molecular architecture of actin cytoskeleton and filopodial growth. Nanoscale Res Lett 2014;9(1):564. doi: 10.1186/1556-276X-9-564. eCollection 2014. PubMed PMID: 25386101; PubMed Central PMCID: PMC4217708.
  147. 147. Pandis C, Trujillo S, Matos J, Madeira S, Ródenas-Rochina J, Kripotou S, Kyritsis A, Mano JF, Gómez Ribelles JL: Porous polylactic acid-silica hybrids: preparation, characterization, and study of mesenchymal stem cell osteogenic differentiation. Macromol Biosci 2015;15(2):262–274. doi: 10.1002/mabi.201400339. Epub 2014 Oct 10. PubMed PMID: 25303745.
  148. 148. Zhou P, Cheng X, Xia Y, Wang P, Zou K, Xu S, Du J: Organic/inorganic composite membranes based on poly(L-lactic-co-glycolic acid) and mesoporous silica for effective bone tissue engineering. ACS Appl Mater Interfaces 2014;6(23):20895–20903. doi: 10.1021/am505493j.
  149. 149. Kim H, Kim I, Choi HJ, Kim SY, Yang EG: Neuron-like differentiation of mesenchymal stem cells on silicon nanowires. Nanoscale 2015;7(40):17131–17138. doi: 10.1039/c5nr05787f. PubMed PMID: 26422757.
  150. 150. Ai H, Lvov YM, Mills DK, Jennings M, Alexander JS, Jones SA: Coating and selective deposition of nanofilm on silicone rubber for cell adhesion and growth. Cell Biochem Biophys 2003;38(2):103–114. PubMed PMID: 12777710.
  151. 151. Yue Z, Liu X, Molino PJ, Wallace GG: Bio-functionalisation of polydimethylsiloxane with hyaluronic acid and hyaluronic acid--collagen conjugate for neural interfacing. Biomaterials 2011;32(21):4714–4724. doi: 10.1016/j.biomaterials.2011.03.032.
  152. 152. Minev IR, Moshayedi P, Fawcett JW, Lacour SP: Interaction of glia with a compliant, microstructured silicone surface. Acta Biomater 2013;9(6):6936–6942. doi: 10.1016/j.actbio.2013.02.048. Epub 2013 Mar 14. PubMed PMID: 23499849.
  153. 153. Patel KR, Tang H, Grever WE, Simon Ng KY, Xiang J, Keep RF, Cao T, McAllister JP II: Evaluation of polymer and self-assembled monolayer-coated silicone surfaces to reduce neural cell growth. Biomaterials 2006;27(8):1519–1526. Epub 2005 Sep 19.
  154. 154. Huang H, Chen L, Wang H, Xiu B, Li B, Wang R, Zhang J, Zhang F, Gu Z, Li Y, Song Y, Hao W, Pang S, Sun J: Influence of patients' age on functional recovery after transplantation of olfactory ensheathing cells into injured spinal cord injury. Chin Med J (Engl) 2003,116:1488–1491.
  155. 155. Lima C, Pratas-Vital J, Escada P, Hasse-Ferreira A, Capucho C, Peduzzi JD: Olfactory mucosa autografts in human spinal cord injury: a pilot clinical study. J Spinal Cord Med 2006;29:191–203; discussion 204–206.
  156. 156. Park HC, Shim YS, Ha Y, Yoon SH, Park SR, Choi BH, Park HS: Treatment of complete spinal cord injury patients by autologous bone marrow cell transplantation and administration of granulocyte-macrophage colony stimulating factor. Tissue Eng 2005;11:913–922.
  157. 157. Yoon SH, Shim YS, Park YH, Chung JK, Nam JH, Kim MO, Park HC, Park SR, Min BH, Kim EY, Choi BH, Park H, Ha Y: Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: phase I/II clinical trial. Stem Cells 2007;25(8):2066–2073.
  158. 158. Chowdary PD, Che DL, Cui B: Neurotrophin signaling via long-distance axonal transport. Annu Rev Phys Chem 2012;63:571–594. doi: 10.1146/annurev-physchem-032511-143704. Epub 2012 Jan 30. Review. PubMed PMID: 22404590.
  159. 159. Chowdary PD, Che DL, Kaplan L, Chen O, Pu K, Bawendi M, Cui B: Nanoparticle-assisted optical tethering of endosomes reveals the cooperative function of dyneins in retrograde axonal transport. Sci Rep 2015;5:18059. doi: 10.1038/srep18059. PubMed PMID: 26656461; PubMed Central PMCID: PMC4674899.
  160. 160. Dono R: Fibroblast growth factors as regulators of central nervous system development and function. Am J Physiol Regul Integr Comp Physiol 2003;284(4):R867–R881. Review. PubMed PMID: 12626354.
  161. 161. Hendry IA, Belford DA: Lack of retrograde axonal transport of the heparin-binding growth factors by chick ciliary neurones. Int J Dev Neurosci 1991;9(3):243–250. PubMed PMID: 1718148.

Written By

Sherif M. Amr

Submitted: 21 October 2015 Reviewed: 12 May 2016 Published: 17 August 2016