Spinal Cord Regeneration

3 Spinal Cord Regeneration


Charles H. Tator


There are two broad categories of strategies to promote repair of the injured spinal cord: neuroprotection and regeneration (Table 3.1). To date, neuroprotection has not produced major recovery after spinal cord injury (SCI); treatment with agents such as steroids or antiinflammatory agents has been disappointing, especially for injuries of major severity, such as American Spinal Injury Association (ASIA) grade A injuries, with complete absence of voluntary motor function and sensation below the level of the injury. Even for less severe injuries, neurologic improvement in patients treated with neuroprotective agents has been minimal1 and not without controversy.2 Ideally, clinicians would like to have a variety of effective neuroprotective agents to counteract the many secondary injury mechanisms that are known to cause progressive damage to the injured spinal cord. Furthermore, it would be highly desirable to have effective neuroprotective strategies for patients with injuries of all severities. The secondary injury mechanisms that require treatment include ischemia, inflammation, excitotoxicity, major electrolyte shifts, and apoptosis. Patients with less severe injuries would benefit if these agents prevented further damage to the spinal cord, and patients with the most severe injuries would benefit from limiting the extent of the damage.


The pathologic effects of acute SCI spread centrifugally in all directions to cause ventral-dorsal, lateral, and rostrocaudal damage.3 Ideally, effective neuroprotective agents would limit the pathologic spread and shorten the distance that regenerating axons would have to travel rostrocaudally in the cord. It is well known that the end result of this cascade of pathophysiologic events is major necrosis and cavitation, a process that has been termed posttraumatic infarction of the cord.3,4 The cavity that forms can be up to several centimeters long rostrocaudally, and thus large portions of spinal cord tissue would have to be replaced by actual ingrowths from the stumps or replacement by transplantation to improve function. In general, it is highly likely that improvement of neurologic recovery after major cord injuries would require a combination of regenerative strategies designed to produce regeneration or replacement of lost tissue. The regenerative strategy that is most likely to be successful is transplantation of cells or tissues, but it is unlikely that this will be sufficient. The goal of functional recovery will require a combination of strategies that include neurotrophic factors and agents to minimize scarring and to counteract endogenous inhibitors that are known to be plentiful in the injured cord.1 Also, combination therapy must include attention to rehabilitative measures such as gravity-assisted ambulation, and some of these rehabilitative measures have proven effectiveness.5,6



Table 3.1 Two Categories of Strategies Promoting Spinal Cord Repair






















Neuroprotection • Has not produced major recovery after spinal cord injury (SCI)

• Includes treatments involving agents such as steroids or antiinflammatory agents

• Ideally, future use may protect patient from secondary injury such as ischemia, inflammation, excitotoxicity, electrolyte shifts, and apoptosis and prevent necrosis and cavitation

• Agents limit pathologic spread of injury and shorten distance for regenerating axons
Regeneration • Likely that regeneration is needed for improvement in neurologic recovery

• Strategy with greater chance of success is the transplantation of cells or tissues

• Combination of strategies likely to be required

 


image The Treatment Window at Various Stages of Spinal Cord Injury


Neuroprotective strategies such as surgical decompression, are designed to counteract the secondary mechanisms of injury. Each strategy has a specific time course and therapeutic window for effectiveness.7 It is also apparent from experimental studies that regenerative strategies are subject to therapeutic time windows. For example, the acutely injured spinal cord is less amenable to the survival of transplanted cells. There is greater survival of transplanted cells in the subacute stage.811 However, the chronic stage may be the most difficult with respect to improving neurologic function because of the long distances that axons would have to regenerate. For example, axons continue to die back for several months after SCI. Also, the classic view is that the astrocytic scarring that develops progressively in the subacute and chronic stages will hinder axonal regeneration, although this is still controversial.



Table 3.2 Specific Mechanisms to Enhance Regeneration after Spinal Cord Injury


































Promotion of endogenous regeneration Includes:

• Regeneration of axons

• Collateral sprouting of axons

• Renewal of astrocytic or oligodendrocytic cells

• Blood vessels by angiogenesis

• Neuronal somata
Regeneration by replacement with transplantation Includes:

• Oligodendrocytes or Schwann cells for myelination

• Astrocytes for axonal guidance

• Neurons to act as “relay stations”

• Synthetic channels for axonal guidance and prevention of scarring

• Hematopoietic cells

• Microglia/macrophages

 


image Types of Regeneration


There are a two main types of regeneration that can be accomplished (Table 3.2). The first depends on regrowth of endogenous cells, and here there are many cell types that need to be considered. The more severe the lesion, the greater the number of cell types that will have to be regrown to accomplish restoration of function. For example, with mild lesions, regeneration of axons or sprouting of collaterals may suffice, whereas more severe lesions may require regeneration of axons, glia, and blood vessels. Regeneration of neuronal cell bodies is possible, but it is extremely unlikely that they would establish useful functional relationships. Similar considerations relate to the specific aims of regeneration, as indicated in Table 3.3. It may be feasible to devise strategies to replace several cell types, and this may be possible with stem/progenitor cell transplantation.


With respect to axonal regeneration and collateral sprouting, there are several anatomical sites where this can occur, as shown in Fig. 3.1. Axonal regeneration is defined as occurring from the injured axon itself, either from the damaged end or from a more proximal location on the damaged axon. In contrast, collateral sprouting occurs from an undamaged axon, possibly a neighboring axon in the same tract or from an axon in a different tract. The collateral may grow at the injury site or from a remote site. As well, a new neuron at the origin of the tract in the cerebral cortex or brainstem may give rise to a replacement axon.



Table 3.3 Aims of Regenerative Strategies
















Regrowth of surviving stumps • Axonal regeneration: regrowth from damaged axon (axonal regeneration is more difficult but may result in more accurate functional connections)

• Sprouting: regrowth from a nearby intact axon
Revascularization of restored tissue • Prevent ischemia

• Use of fibroblast growth factor-1 (FGF-1)

 


image

Fig. 3.1 Types of regeneration and regrowth after spinal cord injury (SCI), which may involve regeneration from the original damaged axon, at or remote from the injury site, or from another neuronal soma at the origin of the tract in the cerebral cortex or brainstem. In contrast, collateral sprouting is from a neighboring undamaged axon at or remote from the injury site. (A) Normal brain and spinal cord showing an axon from the corticospinal tract synapsing with an anterior horn cell in the lumbar cord. (B) Thoracic spinal cord injury (shaded area) has completely interrupted the axon, which then shows caudal wallerian degeneration. (C) Axonal regeneration from the original damaged axon. (D) Axonal regeneration from a rostral segment of the originally damaged axon in the thoracic cord. (E) Axonal regeneration from a proximal segment in the brainstem of the originally damaged axon. (F) Collateral sprouting from an adjacent undamaged axon of another tract, such as the rubrospinal tract, which then grows caudally to synapse with the original denervated anterior horn cell. (G) Collateral sprouting from an adjacent neuron in the cerebral cortex giving rise to an axon of the corticospinal tract, which then regenerates to synapse with the original denervated anterior horn cell. (H) Transplantation of a neuron into the injury site, which then gives rise to an axon that synapses with the original denervated anterior horn cell.


image Aims of the Regenerative Strategies


The necrotic tissue or cavity left behind after acute and subacute/chronic injuries, respectively, have to be replaced with functioning living tissue, and there are many strategies that have been designed in an attempt to accomplish this. Regrowth of tissue from the surviving stumps has been attempted in a variety of ways (Table 3.3). One such way is the promotion of axonal regeneration, which is described as regrowth from the stump of the original transected or otherwise damaged axon. This can be contrasted with sprouting, which is classified as regrowth of an axon from a nearby intact axon. The former is much more difficult to accomplish but might result in more accurate functional improvement. After very severe injuries that transect all the axons present at the site of injury, regeneration from transected axons and transplantation are the only ways to regenerate axons. Regrowth of the supporting tissue including astrocytes and oligodendrocytes would be possible from the stumps, although these cells do not appear to be very mobile, and even if they multiplied in the stumps, as astrocytes are known to do, it is unlikely that they could migrate the long distances necessary to bridge the two stumps. Thus, restoration of glial tissue may have constraints similar to axons, in that replacement by transplantation may be necessary to bridge the large cavities after major SCI.


It is also necessary to consider revascularization of the restored tissue because SCI causes major ischemia of the injured segment, and therefore it would be highly desirable to be able to stimulate regrowth of the vasculature in the area of injury (Table 3.3). Some agents, such as fibroblast growth factor-1 (FGF-1), promote angiogenesis. Other older, unproven strategies such as omental transplantation to restore blood supply12 are not reviewed in detail here.


image Overview and Classification of the Types of Regenerative Strategies for Spinal Cord Injury


Table 3.4 shows the broad variety of focused regenerative strategies that have been studied in experimental models of SCI. Many of these strategies have also been tried in patients, including neurotrophic factors, cell and tissue transplantation, electrical stimulation, and neutralization of inhibitors. Most of the studies in experimental models of SCI have been in the acute and subacute stages, although some also have been in the chronic stage. The studies in patients have been mainly in the subacute and chronic stages and have mostly been phase 1 trials. There have been several comprehensive reviews of the experimental strategies8,1315 and several reviews of the clinical strategies.1,1618 Most of the recent clinical trials of regeneration have been phase 1 trials of the transplantation of cells or tissues involving patients in the subacute and chronic stages of SCI. Unfortunately, very few of these trials have been published in peer-review journals and are not available in any other format in the public domain. Issues regarding design of cell transplantation trials for central nervous system (CNS) diseases in general have been critically reviewed by Cesaro19 and commented on by Sagen.14 The need for great care in translation is evident from the study of Hofstetter et al,20 who showed allodynia-like hypersensitivity of the forepaws due to aberrant axonal sprouting in rats after transplantation of adult neural stem cells into the injured thoracic spinal cord. Furthermore, Dobkin et al21 recently issued a warning about poor trial design, complications, and lack of effectiveness of transplanted olfactory ensheathing glia (OEG) cells in seven patients treated in one very active transplant center. The present chapter provides the experimental background for 25 main strategies and a brief description of any clinical trials of specific strategies.



Table 3.4 Classification of Types of Regenerative Strategies











































































































Neurotrophic factors • Nerve growth factor (NGF)

• Brain-derived growth factor (BDGF)

• Neurotrophin 3 (NT-3)

• Fibroblast growth factor-1 or -2 (FGF-1 or -2)

• Epidermal growth factor (EGF)

• Many others
Matrices and scaffolds • Collagen

• Matrigel

• Millipore

• Synthetic materials (hydrogels, polyesters)

• Natural materials (chitosan)
Neuronal/axonal growth stimulation • Cyclic adenosine monophosphate

• Gangliosides

• Inosine
Cell and tissue transplantation • Spinal cord segments

• Peripheral nerve grafts

• Schwann cells

• Macrophages

• Porcine stem cells

• Blood stem cells

• Bone marrow stromal stem cells

• Umbilical cord/blood stem cells

• Olfactory ensheathing glia or bulb stem cells

• Human embryonic stem cells

• Many others
Stimulation of endogenous stem cells • Growth factors

• Erythropoietin
Electrical stimulation • Direct current fields

• Alternating current
Reduction of scarring • Enzymes (chondroitinase ABC)
Manipulation of extracellular matrix • Tenascin

• Many others
Neutralization of inhibitors • Immunotherapy with antibodies

• Anti-Nogo antibodies

• Nogo receptor antagonists

• Rho inhibition
Fusion molecules • Polyethylene glycol
Manipulation of signaling molecules • Ephrins

• Semaphorins
Rehabilitation strategies • Exercise

• Functional electrical stimulation (FES)

• Gravity-assisted ambulation
Immunomodulation • Immunization

• Macrophages

 


Space does not permit detailed description of some of the other strategies listed in Table 3.4.


Neurotrophic and Growth Factors (Table 3.5)


Neurotrophic and growth factors have been major strategies in experimental SCI.22,23 Many of these agents have produced remarkable growths of proliferating glial, fibrous, and Schwann cells. In our laboratory, direct injection of brain-derived neurotrophic factor (BDNF) produced the most remarkable proliferation of cells at the injury site, with virtually complete disappearance of the cavitation.24 To date, these agents have not been used in SCI patients, except for the Cheng et al25 application of FGF-1 in patients in whom peripheral nerve grafts were transplanted into the spinal cord. However, two of these agents have been used in patients with amyotrophic lateral sclerosis (ALS). Ciliary neurotrophic factor (CNTF) was delivered intrathecally through an indwelling catheter attached to a pump in a phase 1 trial in four patients with ALS. There was no improvement, but there were complications and side effects.26 An initial study with BDNF delivered subcutaneously in ALS patients was not beneficial, and the subsequent trial of BDNF by intrathecal catheter and pump was a phase 3 randomized prospective controlled trial organized by Amgen,27 but the final result has not been reported.



Table 3.5 Neurotrophic and Growth Factors






















Brain-derived neurotrophic factor (BDNF) • Produced the most remarkable cell proliferation at site of injury

• Virtually complete disappearance of cavitation

• To date, not used in SCI patients (except FGF-1 in Cheng study)

• Used in amyotrophic lateral sclerosis (ALS) patients; delivered subcutaneously and no benefits seen
Ciliary neurotrophic factor (CNTF) • Used in ALS patients

• Delivered intrathecally via catheter

• No improvement; complications and side effects present

Note: These factors have been major strategies in experimental SCI and have shown growth of glia, fibroblastic, and Schwann cells.


 


Manipulation of Endogenous Spinal Cord Stem Cells (Table 3.6)


The use of endogenous stem cells for regeneration of the spinal cord after SCI is a highly attractive method for enhancing regeneration of the spinal cord that theoretically could result in the regrowth of new glial cells and new neurons. This strategy is based on the relatively recent discovery by Reynolds and Weiss28 in 1996, which noted the presence of endogenous stem cells in the adult mammalian spinal cord. This has now been confirmed in several laboratories, including mine, and has led to extensive experimentation on methods of stimulating these stem cells to proliferate, differentiate, and migrate into the injured area after SCI. One of the major problems is that the most multipotential cells are located in the ependyma region around the central canal, whereas those located in the parenchyma of the cord are progenitors with less potential.29 Central hemorrhagic necrosis and posttraumatic infarction are cardinal features of the pathology of acute SCI and cause the destruction of the stem cells at the epicenter of the lesion and rostrocaudally for a considerable distance.3,30 Most of the efforts to enhance the proliferation and migration of endogenous stem cells have been with growth factors such as epidermal growth factor (EGF) and FGF-2 and have been successful in increasing the proliferation of these cells but have resulted in only minimal improvement in functional recovery in experimental SCI.29 There have also been efforts to enhance survival of these endogenous stem cells after injury, but mainly in brain injury.31


GM1 Ganglioside (Table 3.7)


GM1 ganglioside is present in neuronal membranes and was studied extensively in animal models of SCI and CNS ischemia, as reviewed by Geisler et al.32 It is of interest that this agent also has neuroprotective effects. After an initial positive result in a small series of patients, a subsequent multicenter randomized prospective control trial in approximately 800 patients failed to show an overall significantly improved neurologic recovery, although the authors pointed to some improvement in the ASIA grade B incomplete injuries.33,34 This was a landmark study and provided a great deal of useful information about the epidemiology and natural history of neurologic recovery in patients with SCI.



Table 3.6 Endogenous Stem Cell Usage for Regeneration















Logic Theoretically, can enhance regrowth of glial cells and new neurons, as precursors of these stem cells (progenitors) were discovered in adult mammalian spinal cord
Problems Multipotential cells are located in ependyma regions around central canal (those in parenchyma of cord have less potential); necrosis and infarction associated with SCI will kill these cells at injury site, with some spread rostrocaudally
Growth factors EGF and FGF-2 have been used successfully to increase proliferation of stem cells; however, functional recovery minimal

 


Transplantation of Whole Spinal Cord Segments (Table 3.8)


Reier et al35 pioneered the implantation of fetal spinal cord in experimental SCI, and there have been some remarkable examples of the potential for this method in experimental studies. One of the best examples was produced by Iwashita et al,36 who transplanted fetal spinal cord into the completely transected spinal cord of newborn rats and showed impressive fusion of the transplant with the adjacent spinal cord stumps and excellent neurologic recovery. Patients with syringomyelia in Sweden37 and the United States38 were treated with implants of human fetal spinal cord in an attempt to obliterate the cavities and stop the progression of neurologic deterioration. It appears that the procedure can be performed without major complications, but the survival of the grafts and long-term benefit have not been proven.



Table 3.7 GM1 Ganglioside in Regeneration



















Logic • May be useful as it is located in neuronal membranes

• May have neuroprotective effects as well
Geisler et al’s randomized prospective controlled trial (RPCT) • No overall improvement in neurologic recovery

• Some improvements in ASIA grade B, incomplete SCI patients
Relevance This study provided information on epidemiology and natural history of neurorecovery in SCI patients

 


Transplantation of Autologous Activated Macrophages (Table 3.9)


Schwartz et al39 in Israel studied the effect of transplanting into spinal cords, and found homologous macrophages from the peripheral blood that had been incubated with peripheral nerves and found improved recovery in rats with experimental SCI. In a phase 1 trial in patients, autologous activated macrophages prepared from the peripheral blood of patients with SCI were incubated in tissue culture conditions in the presence of autologous skin, then injected at surgery into the injured spinal cord just caudal to the epicenter of the injury within 14 days of injury. Three of eight ASIA grade A thoracic cases showed improved neurologic recovery to ASIA grade C,40 and a second publication about this strategy in 2005 reported that 5 of 14 patients treated had neurologic improvement.41 A further trial is in progress.



Table 3.8 Fetal Spinal Cord Implantation Method for Regeneration
























Washita et al study • Transplanted fetal spinal cord into completely transected stem cells of infant rats

• Results:

  1. Fusion with adjacent stumps

  2. Excellent neurorecovery
Treatment for syringomyelia • Goal: obliterate cavities and halt neurologic deterioration with transplants of human fetal spinal cord

• Results:

  1. Procedure done without complications

  2. Survival of grafts and long-term benefits not yet proven

 


Peripheral Nerve Grafts (Table 3.10)


Several clinical applications of peripheral nerve grafts have been reported based on the initial animal experiments of Richardson et al’s group.42 Cheng et al43 then showed remarkable recovery in rats after complete transection of the thoracic spinal cord. In Olson’s laboratory in Sweden, this group inserted peripheral nerves and FGF-1 in fibrin glue to bridge from white to gray matter.43 Cheng et al25 then applied this strategy to a patient in Taiwan with a chronic SCI from a stab wound at 4 years after injury and reported recovery of neurologic function; other patients have also been treated with this strategy in Taiwan (Cheng, personal communication). Peripheral nerve grafts have also been used in eight patients from Brazil without apparent benefit.44 In one patient, Brunelli and von Wild45,46 in Italy inserted autologous sciatic nerves into the corticospinal tract of the spinal cord rostral to the injury site and then anastomosed them to the femoral nerves to restore quadriceps function and directly to the gluteus muscles several months after injury; then reported improvement. Tadie et al47 in France reported a similar case in which peripheral nerve grafts bridged from the cord rostral to the injury site to the L2-L4 spinal roots. Several years earlier, Carlstedt et al48 in Sweden reported recovery of upper limb function after brachial plexus injuries treated with implantation of the avulsed nerve roots directly into the spinal cord. In 10 patients with root reinsertion from 10 days to 9 months after injury, the results were beneficial in at least three cases. Thus, there is some evidence that peripheral nerve bridging is worth pursuing.


Table 3.9 Autologous Activated Macrophages in Regeneration


















Autologous activated macrophages • Autologous macrophages from peripheral blood, incubated with peripheral nerves of skin
Phase I trial (phase 2 trial started and then discontinued) • Autologous macrophages injected caudal to injury site within 14 days of injury

• Results:

  1. Three of eight ASIA grade A thoracic cases showed improved neurologic recovery

  2. Five of 14 (in another publication) patients had neurologic improvements

 


Transplantation of Bone Marrow Stem Cells (Table 3.11)


Whole bone marrow contains a mixture of hematopoietic cells, various mononuclear cells such as macrophages, and marrow stromal cells. The initial reports in animal studies showed that populations of bone marrow-derived stem cells showed neuronal and glial differentiation and improved neurologic recovery after transplantation into the injury site after SCI.49 These studies generated a large number of other animal trials in experimental SCI models,50 and now also in patients in several countries. The major attractions of this strategy are the ease of obtaining autologous tissue for transplantation and the possibility that bone marrow cells have a “homing instinct” so that they may be effective after administration remote from the injury site, through either intravascular or intrathecal routes. In 2005, Park et al51 in South Korea reported that whole bone marrow transplantation into the injury site produced improvement in ASIA grades in all five patients in a phase 1 trial who received whole bone marrow transplants into the injured spinal cord within 7 to 14 days of injury. All five patients also received intravenous injections of granulocyte-macrophage colony-stimulating factor (GM-CSF), whereas a sixth patient who also recovered received only the latter. There are other trials of this strategy in China, the Czech Republic, and Russia, as noted in my recent review.1


Transplantation of Peripheral Blood Stem Cells (Table 3.11)


For reasons similar to those underlying the use of bone marrow-derived stem cells, peripheral blood-derived stem cells have been used in patients with spinal cord diseases. Janson et al52 reported a trial of intrathecal administration in patients with ALS, but there were few details. There are also unreported trials of this strategy in Brazil and Russia.



Table 3.10 Peripheral Nerve Graft Studies























Cheng et al43 • Inserted peripheral nerves and FGF-1 in fibrin glue, bridging white to gray matter in rats with neurorecovery

• Neurologic recovery after stab wound in one patient with peripheral nerve grafts into cord
Barros et al44 • Nerve grafts into spinal cord showed no benefits
Brunelli and von Wild • Autologous sciatic nerve inserted in corticospinal tract and anastomosed to femoral nerve and gluteus muscles; showed improved muscle function
Tadie et al47 • Peripheral nerve graft bridging from spinal cord rostral to injury to L2-L4 roots; improved muscle function
Carlstedt et al48 • Implantation of avulsed nerve roots directly into spinal cord; recovery of upper limb function following brachial plexus injury

 


Umbilical Cord Stem Cells (Table 3.11)


Currently, homologous umbilical cord blood stem cells are being transplanted into patients with chronic SCI in China. The transplantations are performed intravenously, intrathecally, or directly into the spinal cord. There are some published trials of the use of human umbilical cord stem cells in experimental SCI that have shown some benefit.53,54



Table 3.11 Stem Cell Transplantation











































Bone marrow stem cells Animal study reports • Neuronal and glial differentiation and neurologic recovery following bone marrow stem cell implants into SCI site

Advantages • Ease of obtaining autologous tissue


• “Homing instinct” of marrow cells


• Administer intravascularly or intrathecally

Park et al study • Improvements in ASIA grades in patients receiving marrow stem cell transplants within 7 to 14 days of injury


• Patients also received granulocyte-macrophage colony-stimulating factor (GM-CSF)
Peripheral blood stem cells Janson et al study • Intrathecal administration to ALS patients


• Few details of study found
Umbilical cord stem cells China study • Homologous umbilical cord stem cell transplants in chronic SCI patients


• Performed intravenously, intrathecally, or directly into spinal cord


• Reportedly showed benefits in some published trials; no definite proof
Fetal porcine stem cell xenotransplantation
• Diacrin Company performed trial, injecting pig stem sells directly into human spinal cord; no publication of results
Human embryonic stem cells
• At least one study in United States with no reports yet; one planned trial of particular interest involves differentiation in vitro of human embryonic cell line to an oligodendrocytic phenotype with functional competence for myelination, which could be very beneficial in SCI patients where demyelination is extensive

 


Transplantation of Olfactory Ensheathing Glia and Olfactory Bulb (Table 3.12)



Table 3.12 Olfactory Ensheathing Glia and Olfactory Bulb Transplantation




















Advantages • Autologous tissue obtained easily for use in SCI patients

• Has shown benefits in experimental models
Huang et al57 studies • Use of human fetal olfactory ensheathing glia (OEG) or olfactory bulb transplanted into spinal cord at open surgery

• Guest et al58 reported rapid recovery of one case

• Dobkin et al21 criticized trial design and recorded subsequent cases of serious complications, including meningitis

• More trials in progress; incomplete reporting of results and poor trial design

 


Transplantation of OEG is currently being performed in patients with SCI in several countries, including China, Portugal, Russia, and Australia, and is based on many favorable reports in experimental models.55,56 The strategy is attractive because of the ease with which autologous tissue can be obtained for transplantation in patients with SCI. Huang et al57 in China has had the largest experience with this technique, and in 2003, they reported the results in 171 patients, mostly in the chronic phase, in a case series report. They transplanted OEGs or olfactory bulb tissue from human fetuses into the spinal cord at open surgery. Unfortunately, there was no blinding of examiners and no randomization, and neurologic improvement was reported after follow-up of only 8 weeks. Recently, Huang reported that he has performed transplantation of autologous OEG cells into the spinal cord in more than 300 SCI patients (personal communication). Guest et al58 reported rapid recovery of one of Huang’s cases but could not determine the exact mechanism. As noted above, Dobkin et al21 criticized this trial on the basis of poor trial design, and they recorded complications, including meningitis. These authors also recorded the absence of neurologic improvement in all seven patients who had objective neurologic assessment before and after transplantation of the OEG cells. Other investigators in China have also been performing this treatment (personal communication), and a similar large trial of OEG transplantation is in progress in Lisbon, Portugal, by Lima et al,59 who reported the results in seven ASIA grade A patients, all of whom improved in a pilot study that was not a randomized control trial.59 An Australian phase 1 trial of OEG transplants in three patients was reported in 2005,60 and a larger trial is being planned for Australia and New Zealand.


Table 3.13 Schwann Cell Transplantation















Miami Project Has studied Schwann cell strategy for years in experimental animals
Zhu study Injection of Schwann cells from peripheral nerves into spinal cord of SCI patients showed improvements in ASIA and sensory scores
Feng study Injection of Schwann cells from sural nerve into spinal cord of SCI patients showed some recovery

 


Schwann Cell Transplantation (Table 3.13)


The Schwann cell strategy has been pursued in experimental SCI for several decades by many investigators, especially those at the Miami Project. Based on animal studies with human Schwann cells transplanted into the spinal cord of rats with demyelination,61 there have been plans to transplant Schwann cells into the spinal cord of patients with conditions such as multiple sclerosis, but no clinical reports have been published in peer-reviewed journals. Recently, Zhu and colleagues in China injected human fetal Schwann cells harvested from peripheral nerves into the spinal cord of 47 patients with SCI and found improvement in ASIA motor and sensory scores (personal communication). Feng and colleagues in Tianjin, China, injected autologous Schwann cells from sural nerve into the spinal cord of nine SCI patients with apparent recovery in some (personal communication). There is no indication that the examiners were independent or blinded in these apparently phase 1 trials.


Fetal Porcine Stem Cell Xenotransplantation


In the United States, the Diacrin Company trial of fetal stem cells injected directly into the spinal cord involved 10 patients with SCI in two centers, St. Louis and Albany, New York (personal communication), but no report has appeared in the literature.


Human Embryonic Stem Cells


There are a large number of promising preclinical trials of various human embryonic stem cells, some of which have been differentiated toward a neural phenotype.62,63 Some of the initial experimental paradigms in this field were not relevant to humans, such as mouse embryonic stem cells transplanted into rats with SCI.64 However, there has been at least one phase 1 study of human embryonic stem cells in SCI patients in the United States (personal communication), but no published report has appeared. One of the most interesting has been a human embryonic cell line differentiated in vitro to favor an oligodendrocytic phenotype with functional competence for myelination in rats.6567 There is extensive demyelination associated with human SCI, and thus this strategy is very attractive. It is being prepared for a phase 1 trial in SCI (personal communication).


Immunomodulation (Table 3.14)



Table 3.14 Other Strategies


























Immunomodulation • Alter immune response in spinal cord to enhance regeneration

• Done via vaccination or depletion of circulating macrophages by liposome-encapsulated clodronate

• Both cell-mediated and antibody-mediated immune responses can help in axonal regeneration
Fusion molecules (polyethylene glycol) • Evidence that polyethylene glycol can repair neuronal membranes in vitro via “fusion”

• Clinical trial being considered
Cyclic AMP • Injected directly as dibutyryl cAMP or inhibition of cAMP hydrolysis by a phosphodiesterase IV inhibitor (rolipram); can enhance axonal regeneration following SCI
Rho antagonist • Inactivation of Rho via Rho kinase inhibition can promote axonal regeneration after experimental SCI

• Cethrin, a Rho inhibitor applied extradurally at time of surgery; phase 1 trial in patients has been completed with promising results

 


Some of the strategies already discussed, such as transplantation of autologous macrophages, may have an immunomodulation basis, but there have been other specific attempts at altering the immune response in the spinal cord designed to enhance regeneration by vaccination68 or by depletion of circulating macrophages by measures such as liposome-encapsulated clodronate.69 As discussed below, myelin contains molecules that inhibit regeneration of axons, and neutralizing the activity of these inhibitors can enhance axon regeneration. Thus, there are complex CNS-immune system interactions after SCI, and both cell-mediated and antibody-mediated immune responses can help in promoting axonal regeneration. The above efforts to develop an effective vaccine or cytokine treatment for experimental SCI have not yet been examined in human SCI.



Table 3.15 Bioengineering in Neuroprotection and Regeneration

























Examples • Axonal guidance channels

• Subarachnoid drug delivery systems
Advantages • Opportunity to counteract secondary injury

• Provides scaffolds to replace lost tissue and enhance regrowth of axons

• Synaptic plasticity

• Inhibition of astrocytosis

• Structural and chemical versatility
Clinical trials None yet

 


Polyethylene Glycol Application


There is evidence that polyethylene glycol can repair neuronal membranes in vitro by “fusion,” and there has been support for this concept in experimental SCI models.70,71 A clinical trial is being considered (personal communication).


Scaffolds, Matrices, and Other Bioengineering Strategies (Table 3.15)


Bioengineering has been of major importance for exploring new methods of spinal cord repair, and our group has recently reviewed the field, including our experiences with some of these strategies, such as axonal guidance channels and subarachnoid drug delivery systems.72 As stated above, the pathophysiology of SCI is multifactorial and multiphasic; therefore, it is likely that effective treatments will require combinations of neuroprotection and regeneration strategies. Bioengineering offers opportunities to counteract secondary injury, provide scaffolds to replace lost tissue, and enhance axonal regrowth, synaptic plasticity, and inhibition of astrocytosis.73 Biomaterials have major advantages for spinal cord repair because of their structural and chemical versatility. For example, bioengineering technology can facilitate cellular treatment strategies, including Schwann cells,74 olfactory ensheathing glia, or neural stem cells for repair of the injured spinal cord. Currently, the emphasis in our laboratory is on the use of naturally occurring agents, such as chitosan, and to make them biodegradable. To date, none of these strategies has been applied to humans for spinal cord regeneration.


Anti-Nogo-A Inhibition and Nogo-66-Receptor Inhibition (Table 3.16)



Table 3.16 Myelin-Based Protein Inhibitors















Function of myelin-based proteins • Serve as powerful inhibitors of neurite outgrowth in vitro and axonal regeneration in vivo
Examples • Nogo-A, Nogo-66 receptor
Anti-Nogo-A antibody • Counteracts Nogo-A inhibitory protein in vitro and in vivo in rodent and primate studies; phase 1 trial in humans is under way

 


It is now known that there are several myelin-based proteins that are powerful inhibitors of neurite outgrowth in vitro and axonal regeneration in vivo. The most extensively studied of these inhibitors is known as Nogo-A, and it has been cloned and characterized by Schwab and colleagues in Zurich. Nogo-A is a protein present in oligodendroglial myelin and is one of at least three myelin-based inhibitors of central axonal regeneration identified in the CNS. Schwab and colleagues have prepared an antibody to Nogo-A, and this anti-Nogo-A antibody counteracts the Nogo-A inhibitory protein in vitro and in vivo in several experimental models of SCI in various species, including rodents and monkeys.75,76 A “humanized” form of the anti-Nogo-A antibody is currently being administered intrathecally to patients with SCI in a phase 1 trial in Germany and Switzerland in the centers of the European Consortium (see below; personal communication).



Table 3.17 Reducing Glial Scar Formation






















Glial scar • Contains extracellular matrix (ECM) molecules, including chondroitin sulfate proteoglycans (CSPGs), which are inhibitors of axonal growth in vitro
Chondroitinase ABC • Administered intrathecally

• Counteracts inhibitory proteoglycans of ECM
Reports • Several reports from experimental SCI studies show effectiveness

• Some reports show enzyme may even enhance collateral axonal sprouting in central nervous system (CNS) remote from injury
Clinical trials • Likely will be subjected to clinical trials in humans with SCI

 


Other recently described efforts in animal studies to overcome inhibitory factors include antibodies or inhibitors of the Nogo-66 receptor protein.77,78 These agents might be more effective than anti-Nogo-A antibody, which may neutralize only Nogo and not the other myelin-based inhibitors.


Chondroitinase ABC (Table 3.17)


At sites of CNS injury, a glial scar develops, containing extracellular matrix (ECM) molecules, including chondroitin sulfate proteoglycans (CSPGs). CSPGs are inhibitory to axon growth in vitro, and regenerating axons stop at CSPG-rich regions in vivo. Chondroitinase ABC has been administered intrathecally to counteract these inhibitory proteoglycans in the ECM in experimental SCI.79,80 Recently, several other reports have appeared showing the effectiveness of this enzyme on recovery from experimental SCI, including combination strategies with scaffolds and various cellular transplants.8186 Also, it is interesting that this enzyme may enhance collateral axonal sprouting in the CNS remote from the injury site, which may confer additional benefits.87 It is highly likely that this agent will be subjected to clinical trials in humans with SCI.


Cyclic Adenosine Monophosphate


It was first reported in 1991 that cyclic adenosine monophosphate (cAMP) enhances neurite outgrowth in vitro,88,89 and recently it has been shown that this agent can enhance axonal regeneration after experimental SCI.9092 It can be injected directly as dibutyryl cAMP, or cAMP levels can be increased by inhibition of cAMP hydrolysis by the phosphodiesterase IV inhibitor rolipram. This strategy has also been used in combination therapy with neurotrophic factors and with cellular transplants.92 Clinical trials are likely to appear.


Rho Antagonist


McKerracher and colleagues93,94 showed that inactivation of Rho by Rho kinase inhibition can promote axonal regeneration after experimental SCI. Cethrin is a Rho inhibitor and is now undergoing a multicenter phase 1 trial. The agent is applied extradurally at surgery following surgical treatment for decompression or fusion within 14 days of SCI in ASIA grade A cervical or thoracic patients. It is uncertain whether this type of downstream intracellular inactivation of the inhibitory pathways is more effective than inhibition at the axonal membrane as afforded by anti-Nogo-A.


Electrical Stimulation with Direct Current Fields (Table 3.18)


It has been well established that electrical stimulation with direct current (DC) fields enhances the growth and alters the direction of neurite outgrowth in vitro.9597 Many studies of this modality were performed in experimental SCI by Borgens et al98 in a variety of animal models of SCI and with several different forms of electrical stimulation. For many years this strategy was actively pursued in my laboratory but was ultimately abandoned because the therapeutic index was so unfavorable.99,100 A phase 1 trial of pulsed oscillating current in 10 patients was reported by Shapiro et al101 in cervical and thoracic ASIA grade A cases. Stimulation began within 18 days of injury with surgically implanted electrodes, and some patients showed neurologic improvement. Pulsed electrical stimulation of the spinal cord after SCI in humans has also been performed by Xu and Liu in Beijing, China, on more than 100 patients (personal communication).



Table 3.18 Technology and Rehabilitation in Regeneration Enhancement







































Electrical stimulation with direct current fields • Enhances growth and alters direction of neurite outgrowth in vitro

• Many trials done

• Has unfavorable therapeutic index
Functional electrical stimulation (FES) • Stimulation of muscles to augment intact neurologic input or serve as the sole stimuli to produce movements

• Remains questionable whether it plays a role in neurologic recovery

• Treatments require much labor and are expensive
Gravity-assisted ambulation and automated locomotor training • Gravity-assisted ambulation (body weight support) embodies treadmill training

• Can improve walking, especially in incomplete SCIs

• Great need for randomized prospective controlled trial but effectiveness is proven
Electrical stimulation with alternating currents • Some evidence that this can assist in motor recovery; unproven value

• Unknown relation to neurologic recovery
Rehabilitation and exercise strategies • Exercise enhances recovery via regeneration (e.g., treadmill training)

• Does so through elaboration of neurotrophic factors

• Also provides muscle strength and joint mobility

 


Rehabilitation-Exercise Strategies Enhance Regeneration (Table 3.18)


It has been shown that certain physical rehabilitation measures can enhance neurologic recovery by enhancing regeneration, and it is likely that these measures should begin immediately after SCI to derive the maximum benefit. It is now known that physical exercise enhances regeneration of the CNS through the elaboration of neurotrophic factors. Cotman’s group102104 has been at the forefront in defining the physiologic basis of exercise through the elaboration from the CNS of neurotrophic factors such as BDNF. In experimental SCI, the effect of exercise on neurologic function has been firmly established.105,106 Thus, physiotherapy and occupational therapy in patients with SCI not only enhance muscle strength and preserve joint mobility, they also increase neurotrophic factors capable of promoting axonal regeneration. These findings have prompted clinical SCI trials to include early, vigorous, and consistent rehabilitation practices to maximize neurologic recovery. Also, investigators must ensure that defined rehabilitation programs are applied equally across treatment and control groups. In experimental SCI, these goals of enhanced and consistent physical activity are extremely difficult to achieve, but excellent studies have been done in experimental SCI, showing that measures such as treadmill training likely enhance regeneration.107,108


Functional Electrical Stimulation and “Patterned Neural Activity”


Functional electrical stimulation (FES) is accomplished through stimulation of the muscles to augment any intact neurologic input or to provide the sole stimuli to produce muscle contraction to perform a patterned, sequential function, such as walking or peddling a bicycle. The current question being examined is whether patterned neural activity such as provided by FES can play a role in improving neurologic recovery. Also, it is important to examine the mechanism of this modality in experimental models. It is unknown whether recovery is based on axonal regeneration. This treatment modality was applied to Christopher Reeve in a much publicized report purporting to show that his neurologic recovery was attributable to the treatment.109 These treatments are labor intensive and involve large expenditures for equipment, training, and personnel, and thus, they impact significantly on clinical trials. Phase 1 trials of this modality have been reported.110 There is a need for intensive examination of this strategy in experimental SCI.


Gravity-Assisted Ambulation and Automated Locomotor Training


This new modality of treatment of SCI patients has been termed gravity-assisted ambulation or body weight support and generally embodies treadmill training.111 It has been shown that this method of rehabilitation can improve walking after SCI, especially in patients with incomplete injuries.5 A multicenter trial to evaluate this modality has shown that walking is improved in ASIA grade C and D patients.6 Further study by a randomized prospective controlled trial is essential for the same reasons noted above for FES. The precise mechanism for the improvements is not known, but one study with transcranial magnetic stimulation in patients with chronic SCI provided evidence that treadmill training increased locomotor function through an increase in function of the corticospinal tract.112 A multicenter randomized trial commenced in 2003 and has not yet been reported.113 It is conceivable that there is increased axonal sprouting or regeneration, even in chronic cases. Thus, there is a great need for further clinical and experimental studies of this modality.


Electrical Stimulation of the Spinal Cord by Alternating Current


There is a long history of the use of various forms of alternating current for stimulation of the spinal cord in experimental SCI to improve function,114 and there is some evidence that electrical stimulation can assist motor recovery in patients. There is one report of improvement in walking in an ASIA grade C patient with an implanted extradural spinal cord stimulator.115 It is unknown whether the functional improvement is related to neurologic recovery.


image Conclusion


A large number of strategies have been proven to enhance regenerative processes in experimental in vitro and in vivo studies related to regeneration of the spinal cord. Many of these involve elaboration or administration of growth factors, and many involve stimulation of the body’s endogenous regenerative mechanisms. Transplantation of various cells, especially a large variety of stem cells, has shown impressive effects on the pathologic picture. For example, it is possible to produce enough new tissue to completely fill the major cavities left behind after severe SCI. However, functional restoration of complex activities such as grasping and locomotion continue to elude investigators at both the experimental and clinical levels. In clinical SCI, a large number of trials of strategies to enhance regeneration are in progress in many countries, and several hundred SCI patients have been treated with cellular transplantation techniques. Most of the trials have been phase 1 with no controls, no independent examiners, and no reports in peer-reviewed literature or other sources in the public domain. Lack of rigorous attention to trial design and lack of detailed reporting make scientific evaluation impossible. Further, well-designed trials are necessary based on sound experimental SCI study of these strategies in clinically relevant experimental models.


References

1. Tator CH. Review of treatment trials in human spinal cord injury: issues, difficulties, and recommendations. Neurosurgery 2006;59:957–982, discussion 982–957


2. Hurlbert RJ. The role of steroids in acute spinal cord injury: an evidence-based analysis. Spine 2001;26(suppl):S39-S46


3. De Girolami U, Frosch MP, Tator CH. Diseases of the spinal cord and vertebral column. In: Graham DI, Lantos P, eds. Greenfield’s Neuropathology. 7th ed. Regional Neuropathology, vol 1. London: Arnold, 2002:1063–1101


4. Tator CH. Update on the pathophysiology and pathology of acute spinal cord injury. Brain Pathol 1995;5:407–413


5. Ladouceur M, Barbeau H. Functional electrical stimulation-assisted walking for persons with incomplete spinal injuries: longitudinal changes in maximal overground walking speed. Scand J Rehabil Med 2000;32:28–36


6. Wirz M, Zemon DH, Rupp R, et al. Effectiveness of automated locomotor training in patients with chronic incomplete spinal cord injury: a multicenter trial. Arch Phys Med Rehabil 2005;86:672–680


7. Fehlings MG, Sekhon LH, Tator C. The role and timing of decompression in acute spinal cord injury: what do we know? What should we do? Spine 2001;26(suppl):S101-S110


8. Kulbatski I, Mothe AJ, Nomura H, Tator CH. Endogenous and exogenous CNS derived stem/progenitor cell approaches for neurotrauma. Curr Drug Targets 2005;6:111–126


9. Lu J, Ashwell K. Olfactory ensheathing cells: their potential use for repairing the injured spinal cord. Spine 2002;27:887–892


10. Coumans JV, Lin TT, Dai HN, et al. Axonal regeneration and functional recovery after complete spinal cord transection in rats by delayed treatment with transplants and neurotrophins. J Neurosci 2001;21:9334–9344


11. Okano H, Ogawa Y, Nakamura M, Kaneko S, Iwanami A, Toyama Y. Transplantation of neural stem cells into the spinal cord after injury. Semin Cell Dev Biol 2003;14:191–198


12. Clifton GL, Donovan WH, Dimitrijevic MM, et al. Omental transposition in chronic spinal cord injury. Spinal Cord 1996;34:193–203


13. Reier PJ. Cellular transplantation strategies for spinal cord injury and translational neurobiology. NeuroRx 2004;1:424–451


14. Sagen J. Cellular therapies for spinal cord injury: what will the FDA need to approve moving from the laboratory to the human? J Rehabil Res Dev 2003;40(suppl 1):71–79


15. Schwab ME, Bartholdi D. Degeneration and regeneration of axons in the lesioned spinal cord. Physiol Rev 1996;76:319–370


16. Fehlings MG, Perrin RG. The role and timing of early decompression for cervical spinal cord injury: update with a review of recent clinical evidence. Injury 2005;36(suppl 2):B13-B26


17. Tsai EC, Tator CH. Neuroprotection and regeneration strategies for spinal cord repair. Curr Pharm Des 2005;11:1211–1222


18. McDonald JW. Repairing the damaged spinal cord: from stem cells to activity-based restoration therapies. Clin Neurosurg 2004;51:207–227


19. Cesaro P. The design of clinical trials for cell transplantation into the central nervous system. NeuroRx 2004;1:492–499


20. Hofstetter CP, Holmstrom NA, Lilja JA, et al. Allodynia limits the usefulness of intraspinal neural stem cell grafts; directed differentiation improves outcome. Nat Neurosci 2005;8:346–353


21. Dobkin BH, Curt A, Guest J. Cellular transplants in China: observational study from the largest human experiment in chronic spinal cord injury. Neurorehabil Neural Repair 2006;20:5–13


22. Bareyre FM, Schwab ME. Inflammation, degeneration and regeneration in the injured spinal cord: insights from DNA microarrays. Trends Neurosci 2003;26:555–563


23. Ramer LM, Ramer MS, Steeves JD. Setting the stage for functional repair of spinal cord injuries: a cast of thousands. Spinal Cord 2005;43:134–161


24. Namiki J, Kojima A, Tator CH. Effect of brain-derived neurotrophic factor, nerve growth factor, and neurotrophin-3 on functional recovery and regeneration after spinal cord injury in adult rats. J Neurotrauma 2000;17:1219–1231


25. Cheng H, Liao KK, Liao SF, Chuang TY, Shih YH. Spinal cord repair with acidic fibroblast growth factor as a treatment for a patient with chronic paraplegia. Spine 2004;29:E284-E288


26. Penn RD, Kroin JS, York MM, Cedarbaum JM. Intrathecal ciliary neurotrophic factor delivery for treatment of amyotrophic lateral sclerosis (phase I trial). Neurosurgery 1997;40:94–99 discussion 99–100


27. Kalra S, Genge A, Arnold DL. A prospective, randomized, placebo-controlled evaluation of corticoneuronal response to intrathecal BDNF therapy in ALS using magnetic resonance spectroscopy: feasibility and results. Amyotroph Lateral Scler Other Motor Neuron Disord 2003;4:22–26


28. Reynolds BA, Weiss S. Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. Dev Biol 1996;175:1–13


29. Kulbatski I, Mothe AJ, Keating A, Hakamata Y, Kobayashi E, Tator CH. Oligodendrocytes and radial glia derived from adult rat spinal cord progenitors: morphological and immunocytochemical characterization. J Histochem Cytochem 2007;55:209–22


30. Tator CH. Pathophysiology and pathology of spinal cord injury. In: Wilkins RH, Rengachary SS, eds. Neurosurgery. vol 2. New York: McGraw-Hill; 1996:2847–2859


31. Kolb B, Morshead C, Gonzalez C, et al. Growth factor-stimulated generation of new cortical tissue and functional recovery after stroke damage to the motor cortex of rats. J Cereb Blood Flow Metab 2007;27:983–997


32. Geisler FH, Dorsey FC, Coleman WP. Recovery of motor function after spinal-cord injury: a randomized, placebo-controlled trial with GM-1 ganglioside. N Engl J Med 1991;324:1829–1838


33. Geisler FH, Coleman WP, Grieco G, Poonian D. Measurements and recovery patterns in a multicenter study of acute spinal cord injury. Spine 2001;26(suppl):S68-S86


34. Geisler FH, Coleman WP, Grieco G, Poonian D. The Sygen multicenter acute spinal cord injury study. Spine 2001;26(suppl):S87–98


35. Reier PJ, Houle JD, Jakeman L, Winialski D, Tessler A. Transplantation of fetal spinal cord tissue into acute and chronic hemisection and contusion lesions of the adult rat spinal cord. Prog Brain Res 1988;78:173–179


36. Iwashita Y, Kawaguchi S, Murata M. Restoration of function by replacement of spinal cord segments in the rat. Nature 1994; 367:167–170


37. Falci S, Holtz A, Akesson E, et al. Obliteration of a posttraumatic spinal cord cyst with solid human embryonic spinal cord grafts: first clinical attempt. J Neurotrauma 1997;14:875–884


38. Wirth ED III, Reier PJ, Fessler RG, et al. Feasibility and safety of neural tissue transplantation in patients with syringomyelia. J Neurotrauma 2001;18:911–929


39. Schwartz M, Lazarov-Spiegler O, Rapalino O, Agranov I, Velan G, Hadani M. Potential repair of rat spinal cord injuries using stimulated homologous macrophages. Neurosurgery 1999;44:1041–1045, discussion 1045–1046


40. Knoller N, Auerbach G, Fulga V, et al. Clinical experience using incubated autologous macrophages as a treatment for complete spinal cord injury: phase I study results. J Neurosurg Spine 2005;3:173–181


41. Schwartz M, Yoles E. Macrophages and dendritic cells treatment of spinal cord injury: from the bench to the clinic. Acta Neurochir Suppl 2005;93:147–150


42. Richardson PM, McGuinness UM, Aguayo AJ. Axons from CNS neurons regenerate into PNS grafts. Nature 1980;284:264–265


43. Cheng H, Cao Y, Olson L. Spinal cord repair in adult paraplegic rats: partial restoration of hind limb function. Science 1996;273:510–513


44. Barros TE, Oliveira R, Barros EMK, et al. The use of peripheral nerve bridges to spinal cord injury. J Spinal Cord Med 2003;26:S33


45. Brunelli GA. Direct neurotization of muscles by presynaptic motoneurons. J Reconstr Microsurg 2001;17:631–636


46 von Wild KR, Brunelli GA. Restoration of locomotion in paraplegics with aid of autologous bypass grafts for direct neurotisation of muscles by upper motor neurons-the future: surgery of the spinal cord? Acta Neurochir Suppl 2003;87:107–112


47. Tadie M, Liu S, Robert R, et al. 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;19:909–916


48. Carlstedt T, Anand P, Hallin R, Misra PV, Noren G, Seferlis T. Spinal nerve root repair and reimplantation of avulsed ventral roots into the spinal cord after brachial plexus injury. J Neurosurg 2000;93(suppl):237–247


49. Chopp M, Zhang XH, Li Y, et al. Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport 2000;11:3001–3005


50. Hofstetter CP, Schwarz EJ, Hess D, et al. Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc Natl Acad Sci U S A 2002;99:2199–2204


51. Park HC, Shim YS, Ha Y, et al. 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


52. Janson CG, Ramesh TM, During MJ, Leone P, Heywood J. Human intrathecal transplantation of peripheral blood stem cells in amyotrophic lateral sclerosis. J Hematother Stem Cell Res 2001; 10:913–915


53. Nishio Y, Koda M, Kamada T, et al. The use of hemopoietic stem cells derived from human umbilical cord blood to promote restoration of spinal cord tissue and recovery of hindlimb function in adult rats. J Neurosurg Spine 2006;5:424–433


54. Saporta S, Kim JJ, Willing AE, Fu ES, Davis CD, Sanberg PR. Human umbilical cord blood stem cells infusion in spinal cord injury: engraftment and beneficial influence on behavior. J Hematother Stem Cell Res 2003;12:271–278


55. Li Y, Field PM, Raisman G. Regeneration of adult rat corticospinal axons induced by transplanted olfactory ensheathing cells. J Neurosci 1998;18:10514–10524


56. 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


57. Huang H, Chen L, Wang H, et al. 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


58. Guest J, Herrera LP, Qian T. Rapid recovery of segmental neurological function in a tetraplegic patient following transplantation of fetal olfactory bulb-derived cells. Spinal Cord 2006;44:135–142


59. 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 204–196


60. Feron F, Perry C, Cochrane J, et al. Autologous olfactory ensheathing cell transplantation in human spinal cord injury. Brain 2005; 128(pt 12):2951–2960


61. Kohama I, Lankford KL, Preiningerova J, White FA, Vollmer TL, Kocsis JD. Transplantation of cryopreserved adult human Schwann cells enhances axonal conduction in demyelinated spinal cord. J Neurosci 2001;21:944–950


62. Wu P, Tarasenko YI, Gu Y, Huang LY, Coggeshall RE, Yu Y. Region-specific generation of cholinergic neurons from fetal human neural stem cells grafted in adult rat. Nat Neurosci 2002;5:1271–1278


63. Tarasenko YI, Yu Y, Jordan PM, Bottenstein J, Wu P. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells. J Neurosci Res 2004;78:625–636


64. McDonald JW, Liu XZ, Qu Y, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med 1999;5:1410–1412


65. Keirstead HS, Nistor G, Bernal G, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci 2005;25:4694–4705


66. Keirstead HS. Stem cells for the treatment of myelin loss. Trends Neurosci 2005;28:677–683


67. Totoiu MO, Keirstead HS. Spinal cord injury is accompanied by chronic progressive demyelination. J Comp Neurol 2005;486:373–383


68. David S, Ousman SS. Recruiting the immune response to promote axon regeneration in the injured spinal cord. Neuroscientist 2002;8:33–41


69. Popovich PG, Guan Z, Wei P, Huitinga I, van Rooijen N, Stokes BT. Depletion of hematogenous macrophages promotes partial hindlimb recovery and neuroanatomical repair after experimental spinal cord injury. Exp Neurol 1999;158:351–365


70. Shi R, Borgens RB, Blight AR. Functional reconnection of severed mammalian spinal cord axons with polyethylene glycol. J Neurotrauma 1999;16:727–738


71. Borgens RB, Shi R. Immediate recovery from spinal cord injury through molecular repair of nerve membranes with polyethylene glycol. FASEB J 2000;14:27–35


72. Nomura H, Tator CH, Shoichet MS. Bioengineered strategies for spinal cord repair. J Neurotrauma 2006;23:496–507


73. Jimenez Hamann MC, Tator CH, Shoichet MS. Injectable intrathecal delivery system for localized administration of EGF and FGF-2 to the injured rat spinal cord. Exp Neurol 2005;194:106–119


74. Chau CH, Shum DK, Li H, et al. Chondroitinase ABC enhances axonal regrowth through Schwann cell-seeded guidance channels after spinal cord injury. FASEB J 2004;18:194–196


75. Schwab ME. Experimental aspects of spinal cord regeneration. Curr Opin Neurol Neurosurg 1993;6:549–553


76. Schwab ME. Nogo and axon regeneration. Curr Opin Neurobiol 2004;14:118–124


77. Li S, Liu BP, Budel S, et al. Blockade of Nogo-66, myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein by soluble Nogo-66 receptor promotes axonal sprouting and recovery after spinal injury. J Neurosci 2004;24:10511–10520


78. Li S, Strittmatter SM. Delayed systemic Nogo-66 receptor antagonist promotes recovery from spinal cord injury. J Neurosci 2003;23:4219–4227


79. Yick LW, Wu W, So KF, Yip HK, Shum DK. Chondroitinase ABC promotes axonal regeneration of Clarke’s neurons after spinal cord injury. Neuroreport 2000;11:1063–1067


80. Bradbury EJ, Moon LD, Popat RJ, et al. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 2002;416:636–640


81. Huang WC, Kuo WC, Cherng JH, et al. Chondroitinase ABC promotes axonal re-growth and behavior recovery in spinal cord injury. Biochem Biophys Res Commun 2006;349:963–968


82. Houle JD, Tom VJ, Mayes D, Wagoner G, Phillips N, Silver J. Combining an autologous peripheral nervous system “bridge” and matrix modification by chondroitinase allows robust, functional regeneration beyond a hemisection lesion of the adult rat spinal cord. J Neurosci 2006;26:7405–7415


83. Massey JM, Hubscher CH, Wagoner MR, et al. Chondroitinase ABC digestion of the perineuronal net promotes functional collateral sprouting in the cuneate nucleus after cervical spinal cord injury. J Neurosci 2006;26:4406–4414


84. Ikegami T, Nakamura M, Yamane J, et al. Chondroitinase ABC combined with neural stem/progenitor cell transplantation enhances graft cell migration and outgrowth of growth-associated protein-43-positive fibers after rat spinal cord injury. Eur J Neurosci 2005;22:3036–3046


85. 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;22:226–239


86. 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


87. Barritt AW, Davies M, Marchand F, et al. Chondroitinase ABC promotes sprouting of intact and injured spinal systems after spinal cord injury. J Neurosci 2006;26:10856–10867


88. Ono K, Katayama N, Yamagata Y, Tokunaga A, Tsuda M. Morphology of neurites from N18TG2 cell induced by protein kinase inhibitor H-7 and by cAMP. Brain Res Bull 1991;26:605–612


89. Sanna PP, Bloom FE, Wilson MC. Dibutyryl-cAMP induces SNAP-25 translocation into the neurites in PC12. Brain Res Dev Brain Res 1991;59:104–108


90. Spencer T, Domeniconi M, Cao Z, Filbin MT. New roles for old proteins in adult CNS axonal regeneration. Curr Opin Neurobiol 2003;13:133–139


91. Pearse DD, Pereira FC, Marcillo AE, et al. cAMP and Schwann cells promote axonal growth and functional recovery after spinal cord injury. Nat Med 2004;10:610–616


92. Lu P, Yang H, Jones LL, Filbin MT, Tuszynski MH. Combinatorial therapy with neurotrophins and cAMP promotes axonal regeneration beyond sites of spinal cord injury. J Neurosci 2004;24:6402–6409


93. Lehmann M, Fournier A, Selles-Navarro I, et al. Inactivation of Rho signaling pathway promotes CNS axon regeneration. J Neurosci 1999;19:7537–7547


94. Dubreuil CI, Winton MJ, McKerracher L. Rho activation patterns after spinal cord injury and the role of activated Rho in apoptosis in the central nervous system. J Cell Biol 2003;162:233–243


95. Jaffe LF, Poo MM. Neurites grow faster towards the cathode than the anode in a steady field. J Exp Zool 1979;209:115–128


96. McCaig CD. Spinal neurite reabsorption and regrowth in vitro depend on the polarity of an applied electric field. Development 1987;100:31–41


97. Patel N, Poo MM. Orientation of neurite growth by extracellular electric fields. J Neurosci 1982;2:483–496


98. Borgens RB, Blight AR, McGinnis ME. Behavioral recovery induced by applied electric fields after spinal cord hemisection in guinea pig. Science 1987;238:366–369


99. Hurlbert RJ, Tator CH, Theriault E. Dose-response study of the pathological effects of chronically applied direct current stimulation on the normal rat spinal cord. J Neurosurg 1993;79:905–916


100. Hurlbert RJ, Tator CH. Characterization of longitudinal field gradients from electrical stimulation in the normal and injured rodent spinal cord. Neurosurgery 1994;34:471–482 discussion 482–473


101. Shapiro S, Borgens R, Pascuzzi R, et al. Oscillating field stimulation for complete spinal cord injury in humans: a phase 1 trial. J Neurosurg Spine 2005;2:3–10


102. Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002;25:295–301


103. Cotman CW, Engesser-Cesar C. Exercise enhances and protects brain function. Exerc Sport Sci Rev 2002;30:75–79


104. Neeper SA, Gomez-Pinilla F, Choi J, Cotman C. Exercise and brain neurotrophins. Nature 1995;373:109


105. Gomez-Pinilla F, Ying Z, Opazo P, Roy RR, Edgerton VR. Differential regulation by exercise of BDNF and NT-3 in rat spinal cord and skeletal muscle. Eur J Neurosci 2001;13:1078–1084


106. Van Meeteren NL, Eggers R, Lankhorst AJ, Gispen WH, Hamers FP. Locomotor recovery after spinal cord contusion injury in rats is improved by spontaneous exercise. J Neurotrauma 2003;20:1029–1037


107. Edgerton VR, Tillakaratne NJ, Bigbee AJ, de Leon RD, Roy RR. Plasticity of the spinal neural circuitry after injury. Annu Rev Neurosci 2004;27:145–167


108. Engesser-Cesar C, Anderson AJ, Basso DM, Edgerton VR, Cotman CW. Voluntary wheel running improves recovery from a moderate spinal cord injury. J Neurotrauma 2005;22:157–171


109. McDonald JW, Becker D, Sadowsky CL, Jane JA Sr, Conturo TE, Schultz LM. Late recovery following spinal cord injury: case report and review of the literature. J Neurosurg 2002; 97(suppl):252–265


110. Thrasher TA, Flett HM, Popovic MR. Gait training regimen for incomplete spinal cord injury using functional electrical stimulation. Spinal Cord 2006;44:357–361


111. Dietz V, Colombo G. Recovery from spinal cord injury-underlying mechanisms and efficacy of rehabilitation. Acta Neurochir Suppl 2004;89:95–100


112. Thomas SL, Gorassini MA. Increases in corticospinal tract function by treadmill training after incomplete spinal cord injury. J Neurophysiol 2005;94:2844–2855


113. Dobkin BH, Apple D, Barbeau H, et al. Methods for a randomized trial of weight-supported treadmill training versus conventional training for walking during inpatient rehabilitation after incomplete traumatic spinal cord injury. Neurorehabil Neural Repair 2003;17:153–167


114. Wallace MC, Tator CH, Gentles WM. Effect of alternating current stimulation of the spinal cord on recovery from acute spinal cord injury in rats. Surg Neurol 1987;28:269–276


115. Herman R, He J, D’Luzansky S, Willis W, Dilli S. Spinal cord stimulation facilitates functional walking in a chronic, incomplete spinal cord injured. Spinal Cord 2002;40:65–68


< div class='tao-gold-member'>

Stay updated, free articles. Join our Telegram channel

Jul 7, 2016 | Posted by in CRITICAL CARE | Comments Off on Spinal Cord Regeneration

Full access? Get Clinical Tree

Get Clinical Tree app for offline access