73
Views
5
CrossRef citations to date
0
Altmetric
Review

Potential role of stem cells in severe spinal cord injury: current perspectives and clinical data

, , , &
Pages 15-27 | Published online: 25 Sep 2012

Abstract

Stem cell transplantation for spinal cord injury (SCI) along with new pharmacotherapy research offers the potential to restore function and ease the associated social and economic burden in the years ahead. Various sources of stem cells have been used in the treatment of SCI, but the most convincing results have been obtained with neural progenitor cells in preclinical models. Although the use of cell-based transplantation strategies for the repair of chronic SCI remains the long sought after holy grail, these approaches have been to date the most successful when applied in the subacute phase of injury. Application of cell-based strategies for the repair and regeneration of the chronically injured spinal cord will require a combinational strategy that may need to include approaches to overcome the effects of the glial scar, inhibitory molecules, and use of tissue engineering strategies to bridge the lesion. Nonetheless, cell transplantation strategies are promising, and it is anticipated that the Phase I clinical trials of some form of neural stem cell-based approach in SCI will commence very soon.

Introduction

Spinal cord injury (SCI) is a major cause of disability and, at present, there is no universally accepted treatment. The cervical and lumbar regions of the spine are the most commonly affected areas in SCI. The functional decline following SCI is contributed to by both direct mechanical injury and secondary pathophysiological mechanisms that are induced by the initial trauma. These mechanisms initially involve widespread hemorrhage at the site of injury and necrosis of cellular components of the central nervous system (CNS). At later stages of injury, the cord is observed to display reactive gliosis. The actions of astrocytes, as well as numerous other cells in this response, create an environment that is not conducive to axonal regrowth, and the immune system worsens this further.Citation1

The discovery of the potential utility of stem cells in neurological repair and regeneration is an exciting development in neuroscience. Despite advances in medical and surgical care, the current clinical therapies for SCI are largely ineffective. During the last two decades, the search for new therapies has been revolutionized with the discovery of stem cells, which has inspired scientists and clinicians to search for a stem cell-based reparative approach to treat SCI. Stem cells, in the niches around the ependymal layer, proliferate after neuronal loss. This proliferation is triggered by various messenger cascades provoked by the injury. Gliosis in most situations impairs the attempts of axons to regrow and re-establish communications. Cell replacement approaches in the setting of SCI can be used to achieve two broad goals, ie, regeneration, which seeks to replace lost or damaged neurons and induce axonal regeneration or plasticity, and repair, which seeks to replace supportive cells such as oligodendrocytes in order to induce remyelination and prevent progressive myelin loss.Citation2 In the setting of SCI, stem cell therapy could potentially be used to stimulate the endogenous stem cell population to proliferate along neuronal lines or to supplement stem cells in the repair process.Citation3,Citation4 The mature CNS harbors endogenous stem cells which develop into neurons constantly in at least a few areas of the CNS, ie, the subventricular zone, which includes the linings of the lateral ventricles, the subgranular zone of the dentate gyrus, and the central canal of the spinal cord. These cells normally differentiate only into astrocytes and oligodendrocytes in vivo.Citation5,Citation6 There are some factors in the CNS which normally limit or prevent endogenous stem cells from becoming mature neurons.Citation4

After injury, with resultant loss of both nerve cells and cells that provide the myelin for appropriate conduction properties, the obvious solution would be to provide cells that can replace the lost function. A variety of tissues and cells have been used to encourage restoration of function.Citation7 These include stem cells, olfactory ensheathing cells (cells that form the myelin on olfactory nerves), Schwann cells (cells that form the myelin on peripheral nerves), dorsal root ganglia, adrenal tissue, hybridomas, peripheral nerves, or transplanted conduits of Schwann cells, which would serve as a source for chemical and mechanical guidance. It is postulated that these tissues would rescue, replace, or provide a regenerative pathway for injured adult neurons, which would then integrate or promote regeneration of the spinal cord circuitry and restore function after injury.Citation8

The route through which stem cells can be instilled is a major question in cell therapy. Direct injection into the injury site at the time of surgery, delivery through the vascular route by highly selective angiography, through the central canal by instillation into the fourth ventricle, and intrathecal instillation are the possible and frequently used routes for stem cell transplantation in SCI. Of these routes, intrathecal instillation is technically simple and least likely to provoke iatrogenic damage of regenerative potential.

Due to cellular loss, regeneration after SCI is limited, and current approaches to treatment of SCI do not lead to complete cure. Various sources of stem cell transplantation have been shown to replace host neurons, enhance axonal growth, and improve functional recovery successfully in rat models of SCI.Citation9 New approaches in stem cell therapy using neural stem/progenitor cells are widely accepted in the treatment of degenerative diseases for the repair of damaged or lost tissues (). There are several cell sources for stem cell therapy, including fetal spinal cord tissue,Citation10 neuronal stem cells from adult brains, and mesenchymal stem cells from bone marrowCitation11 and other organs, in addition to embryonic stem cells. Human embryonic and adult stem cells both have potential use as cell-based regenerative therapies. Of course they differ in many respects, eg, in the number and differentiated cell types that they can become.

Figure 1 Major stem cell sources, regenerative strategies, and levels of difficulties in the treatment of spinal cord injury.

Figure 1 Major stem cell sources, regenerative strategies, and levels of difficulties in the treatment of spinal cord injury.

Pathophysiology of SCI

To understand the rationale of the recent advances, it is first necessary to review the pathophysiology of SCI. There are four general types of SCI: cord maceration, in which the morphology of the cord is severely distorted; cord lacerations (gun shot or knife wounds); contusion injury, which leads to a central hematomyelia that may evolve into a syringomyelia; and solid cord injury, in which there is no central focus of necrosis as in contusion injury. There are three phases of SCI response that occur after injury, ie, acute, secondary, and chronic injury processes.Citation12Citation14 Experimental models and clinical observations of acute SCI support the concepts of primary and secondary injury, in which the initial mechanical insult is succeeded by a series of deleterious events that promote progressive tissue damage and ischemia. Whereas the primary injury is fated by the circumstances of the trauma, the outcome of the secondary injury may be amenable to therapeutic modulation.

Acute SCI

In the acute phase, which encompasses the moment of injury and extends for the first few days, a variety of parallel pathophysiological processes begins, which can be summarized in two complex phases.Citation15

Primary injury mechanism

The primary injury phase is due to compression or contusion. Primary injury mechanisms include impact plus persistent compression; impact alone with transient compression; distraction; and laceration/transection.Citation13 The first mechanism in SCI involves cord compression, fracture-dislocations, and acute disc ruptures. The second mechanism involves impact alone with only transient compression and distraction. Forcible stretching of the spinal column in the axial plane provides a third mechanism and becomes apparent when distractional forces resulting from flexion, extension, rotation, or dislocation produce shearing or stretching of the spinal cord and/or its blood supply. This type of injury may underlie SCI without radiological abnormality, especially in children where cartilaginous vertebral bodies, underdeveloped musculature, and ligament laxity are predisposing factors.Citation16

The initial mechanical insult tends to damage primarily the central gray matter, with relative sparing of the white matter, especially peripherally, which can be the result of its softer consistency and greater vascularity.Citation17 Further nerve transmission may be disrupted due to microhemorrhages or edema near the injury site.Citation18Citation20 Damage of gray matter occurs within the first hour after injury, whereas the white matter is irreversibly damaged within 72 hours after injury.

Secondary injury mechanism

Following an initial impact after SCI, there is a cascade of downstream events termed “secondary injury”, which culminates in progressive degenerative events in the spinal cord. These secondary injury mechanisms include, but are not limited to, ischemia, inflammation, free radical-induced cell death, glutamate excitotoxicity, cytoskeletal degradation, and induction of extrinsic and intrinsic apoptotic pathways. After SCI, several factors, including glutamate release, lipid peroxidation, and ionic imbalance, contribute to the events, leading to acute local ischemia and secondary degeneration.Citation21 There is emerging evidence that glutamate excitotoxicity plays a key role not only in neuronal cell death but also in delayed post-traumatic spinal cord white matter degeneration. Importantly however, the differences in cellular composition and expression of specific types of glutamate receptors in grey versus white matter require a compartmentalized approach to understand the mechanisms of secondary injury after SCI.

Chronic SCI

In chronic SCI, the wave of secondary cell death, which mainly affects neurons and oligodendrocytes, spreads rostrally and caudally from the site of impact, leading to structural and functional disturbance of the spinal cord.Citation22 Later, secondary mechanisms cause the chronic phase of the SCI, when they produce vasculature and ischemia, glutamatergic excitotoxicity, oxidative cell stress, lipid peroxidation, and inflammation, all of which alone or in concert can trigger apoptosis.Citation21 Mortality rates are highest in the first year after SCI. For patients surviving at least 1 year after traumatic SCI, life expectancy is approximately 90% of normal.Citation23,Citation24 Higher neurological level, severity of injury, and older age at the time of SCI negatively impact survival.

Current treatments and clinical trials for SCI

Pharmacological agents must be given within a narrow window of opportunity to be effective. Although many therapeutic agents show potential promise in animal models, only methylprednisolone has been shown in large, randomized, double-blind human studies to enhance the functional recovery of neural elements after acute SCI.Citation25 Several studies have investigated chronic SCI models using whole tissue grafts and cells of the peripheral nervous system. Transplantation of fetal spinal tissue, fetal brain cortex, olfactory ensheathing cells, peripheral nerve grafts, and Schwann cells after SCI have all been shown to improve locomotor recovery,Citation26Citation30 suggesting that the chronic post-injury period may be a feasible target for repair ().

Table 1 Recent clinical trials of stem cell treatment for SCI listed on http://www.clinicaltrials.gov

Regeneration and replacement of neurons and glia that undergo cell death soon after injury are the main goals of all stem cell-based therapies for SCI. Successful development of stem cell-based therapies for SCI requires more intense work to obtain a better understanding of stem cell differentiation pathways and their survival upon transplantation.Citation25 All stem cell replacement strategies should address these two important problems, and this may be accomplished by improved differentiation protocols for stem cells, transplantation of neural progenitor cells, or by activation of endogenous sources of neural progenitors. However, the ideal source of stem cells for efficient and safe cell replacement has remained a challenging issue that requires more investigation ().

Table 2 Comparison of different stem cell types used in clinical trials for the treatment of spinal cord injury

An autologous transplantation approach for the treatment of SCI eliminates concerns regarding immune rejection and avoids the controversy over embryonic or neonatal sources. However, an autologous approach necessitates sacrificing a peripheral nerve, and despite improvements in amplification techniques, more time is needed before enough cells can be generated for transplantation. Alternative sources of stem cells from postnatal skin or adult bone marrow have recently been pursued and tested after thoracic transectionCitation31 or contusionCitation32 injuries to obviate the need for harvesting a peripheral nerve from a patient. Different sources of stem cells are being exploited for the treatment of spinal cord injury as well as other neurological disorders.

Human embryonic stem cells

Embryonic stem cells are derived from the inner cell mass of mammalian blastocysts and have the ability to proliferate by maintaining both their pluripotency and the ability to differentiate into nearly all cell types, including neuronal and glial fate cells.Citation26,Citation33 Human embryonic stem cells were first derived from human blastocysts by Thomson et al in 1998.Citation27 These cells have an ability to proliferate in long-term cultures while maintaining their pluripotent nature. Therefore, embryonic stem cells are a promising source of differentiated oligodendrocytes and motor neurons for the treatment of SCI. Several reportsCitation34Citation36 have demonstrated the in vitro capacity of human embryonic stem cells to generate neural progenitor cells, including regionally specific neuronal subtypes. Compared with other sources of cell therapy, human embryonic stem cells are one of the most attractive cell sources for spinal cord therapy and this strategy has been validated recentlyCitation34 by the finding that human embryonic stem cell-derived motor neurons can survive and integrate into the spinal cord. In a recent study by Erceg et al,Citation37 transplantation of human embryonic stem cell-derived oligodendrocyte progenitor cells and/or motor neuron progenitors was done in an adult rat with SCI. The study demonstrated the survival, migration, and differentiation of transplanted cells into appropriate cell types without forming teratomas, and improved locomotor function.

Limitations with regard to this source of cells include the lengthy and complex differentiation protocols needed for obtaining neural progenitors from human embryonic stem cells, application of undesired cell types and undefined factors, and also issues regarding the safety of transplantation of human embryonic stem cells in humans, including formation of teratomas following human embryonic stem cell-derived neural cell engraftment.Citation38

Neural stem cells

Neural stem cells can be isolated from various parts of the embryonic and adult CNS, such as the subventricular zone lining the lateral ventriclesCitation39Citation41 and the dentate gyrus of the hippocampus,Citation42,Citation43 although the multipotentiality of the latter remains a contentious issue. Neural stem cells are multipotent and can differentiate into three lineages, ie, neurons, oligodendrocytes, and astrocytes, and can be efficiently propagated in vitro.Citation44,Citation45 The alternative strategy for treatment of SCI is recruitment of endogenous neural stem cells or transplantation of neural stem cells. There are very few reportsCitation46Citation49 that describe the mechanism of integration of neural stem cells and a mechanism to promote functional recovery after SCI. Recently, Hooshmand et alCitation46 have shown that no change occurs in the host microenvironment (lesion size, tissue sparing, glial scar, and expression of proteins such as fibronectin, NG2, versican, GFAP, and PECAM1) after analysis of neural stem cell-engrafted models of SCI. Sources of transplanted neural stem cells, methods of isolation, and preparation of cells prior to implantation seem to be critical in cell survival and integration after implantation.Citation46,Citation50

Neural stem cells are more preferable to human embryonic stem cells for clinical applications because they are safer for cell therapy, given that neural stem cells have less potential to form tumors compared with embryonic stem cells.Citation51 Isolation of pure populations of differentiated cells, inefficient tracking systems, and moderate cell survival after transplantationCitation45,Citation52 are critical challenges that remain when using neural stem cells. Axonal regeneration and extension by cell replacement using either endogenous or exogenous stem cells suffer from various limitations, including formation of glial scars, a lack of neurotrophic factors, inhibitory myelin-associated molecules, and decreased levels of cAMP.Citation53,Citation54

Induced pluripotent stem cells

Induced pluripotent stem cells represent a breakthrough in the field of stem cell biology and provide a promising cell resource for customized patient-specific cell therapies. Induced pluripotent stem cells are very similar to embryonic stem cells in terms of gene expression, chromatin methylation, and embryoid body and viable chimera formation.Citation55 They are capable of differentiating into all cell types, including neurons, glia, neural progenitor cells, and motor neurons.Citation56,Citation57 Furthermore, the derivation of induced pluripotent stem cells using nonviral methodsCitation58 or by chemicals and small molecules,Citation59 such as transcription factors,Citation60 Oct4, Klf4, Sox2, and c-Myc,Citation61 makes this cell replacement strategy very attractive. Nevertheless, this cell type shares disadvantages similar to those of other cell sources. Teratoma formation, aberrant reprogramming, and the presence of transgenes in induced pluripotent stem cell populations are the most concerning obstacles, which should be addressed before their clinical application.Citation62

Bone marrow stromal cells

Bone marrow stromal cells are isolated from the stromal compartment of the bone marrow and are nonteratogenic, with anti-inflammatory and immunomodulatory effects.Citation63,Citation64 These cells also secrete neurotrophic factors and cytokines which make them an attractive source for trophic support in autologous transplantation of endogenous and co-implanted cells at the time of CNS injury. Several studies in rodents have demonstrated that the intraspinal, intrathecal, and systemic (intravenous) routes of delivery have been successful for axonal regrowth and sprouting in SCI repair.Citation65

However, the use of bone marrow stromal cells in SCI is limited because of certain issues, such as bone marrow stromal cell migration beyond the injection site. Variability in clinical outcome of bone marrow stromal cells across the lesion site in SCI repair makes it difficult due to intervariability in efficacy and immunomodulatory potency. Also, there is an absence of more appropriate models for SCI using bone marrow stromal cells to distinguish between functional preservation of axons and de novo axonal regrowth.Citation66,Citation67

Umbilical cord blood-derived stem cells

Identification of a suitable source of stem cells for use in regenerative medicine research and its applications has been challenging. For effective clinical application, a readily, more accessible, abundant, and compatible source of stem cells is required. Umbilical cord blood provides an answer to these problems because it is an abundant source of nonembryonic stem cells.Citation68 Compared with the collection of bone marrow, umbilical cord blood collection is more feasible because it is noninvasive with no side effects for either the baby or the mother.Citation69,Citation70 Moreover, umbilical cord blood stem cells have a higher proliferating potential, with telomeres that are longer than those of other somatic stem cells.Citation71,Citation72 Further, umbilical cord blood can be stored and cryopreserved in cord blood banks for later use in transplant applications.

It has been shown that the mononuclear fraction of umbilical cord blood contains pluripotent stem cells with the potential to become neural cells.Citation73 The use of these cells in neural clinical applications is still in the initial phase, but the results obtained so far are very promising. Kang et alCitation74 purified mesenchymal stem cells from umbilical cord blood and transplanted them to the injury site in a 37-year-old woman with SCI. The results showed regeneration of the spinal cord at the injury site and the patient showed improved sensory perception and mobility.Citation74

Preclinical trials based on umbilical cord blood stem cells in animal models have shown promising results in the treatment of neural diseases and injuries.Citation75 Many researchers have demonstrated that umbilical cord blood stem cells are amenable to neurological application, including preclinical animal models of disease, and more recently clinical trials.Citation76 Therefore, umbilical cord blood stem cells are unique in their suitability for use in stem cell transplantation for the treatment of neurological disease.Citation77

Adipose-derived mesenchymal stem cells

Studies have been conducted with mesenchymal stem cells for the treatment of various diseases in the field of regenerative medicine. For many years, bone marrow stromal cells have been the primary source of stem cells for tissue engineering applications.Citation78Citation80 However, studies have shown that subcutaneous adipose tissue has an advantage over other stem cell sources, ie, the ease of isolating stem cells from harvested tissue.Citation81 Adipose mesenchymal stem cells show stable growth and proliferation kinetics, and can differentiate into adipogenic, osteogenic, chondrogenic, myogenic, or neurogenic lineages in vitro.Citation82Citation84

Adipose mesenchymal stem cells express CD90, CD105, CD106, CD117, and STRO-1 markers, and are negative for hematopoietic lineage markers (eg, CD45 and CD14) as well as for endothelial cell markers (CD31, CD144) and von Willebrand factor.Citation82,Citation84 Adipose mesenchymal stem cells are fibroblast-like cells in their morphology, and preserve their shape after expansion in vitro.Citation82,Citation85,Citation86

Adipose mesenchymal stem cells can be used in the treatment of many CNS injuries.Citation87Citation91 Ryu et alCitation91 used this type of stem cell for the treatment of acute spinal injury in a canine model, and reported significant improvement in neurological function. In another study, adipose mesenchymal stem cells were used to treat SCI in rats after in vitro differentiation into Schwann cells.Citation92 In this study, the authors stated that functional improvement was not observed despite the significant histological improvement achieved at the primary injury site by the transplantation of Schwann cells. Therefore, it was speculated that complete recovery of spinal cord function requires a much more complex treatment strategy combining different modalities. Adipose mesenchymal stem cell therapies appear very promising based on the results of in vitro and in vivo research. Currently, a number of clinical trials for the regeneration of soft tissue, craniofacial tissue, cardiovascular, and CNS tissues are underway.Citation93

Schwann cells

Schwann cells are typical gliocytes that are involved in axon regeneration in the peripheral nervous system and form the myelin sheaths around axons.Citation94 Several neurotrophic factors, extracellular matrix, and cell adhesion molecules expressed by Schwann cells have been used to promote axon regeneration and improve the glial environment after SCI.Citation95,Citation96

It is believed that astroglial scars block the growing axons and prevent migration of Schwann cells into the astroglial domain, so they are unable to provide a bridge for newly born axons to grow through the astroglial scar.Citation95 Therefore, more research is needed to maintain the biological activity of Schwann cells and increase their migration after being transplanted to the SCI site. Woodhoo et alCitation97 implanted Schwann cell precursors into the injured spinal cord and found that these cells have greater ability than normal Schwann cells to migrate and survive in the astroglial environment.

In another study, Papastefanaki et alCitation98 transplanted Schwann cells with altered adhesion properties into an in vivo mouse model of SCI and found that these cells promoted faster and significantly greater functional recovery compared with normal Schwann cells or no cells at all. The ability of Schwann cells to repair SCI can be enhanced by combined therapeutic measures, such as increasing the secretion of neurotrophic factors by genetic engineering, filling the gap, and providing channels through the injury site.

Olfactory ensheathing cells

Olfactory ensheathing cells are present along the full length of olfactory nerves crossing the peripheral nervous system and CNS junction.Citation99,Citation100 Unlike most of the adult CNS, the olfactory bulb retains its regenerative capacity. Compared with other cells, olfactory ensheathing cells have a greater ability to migrate to areas distal from the transplantation site, so they can bridge the gap between the lesion site and normal spinal cord to grow the axons.Citation101 These cells can be harvested and then transplanted back into the original human donor, thereby eliminating rejection-related problems and being ideal for translational research.Citation102

Olfactory ensheathing cell transplantation studies in animals suggest that delaying transplantation after SCI may be beneficial to the survival of ensheathing cells.Citation98 There are many published reports about autologous transplantation as a treatment for SCI.Citation106 Olfactory ensheathing cells have more potential advantages than Schwann cells, but more basic research is needed to identify the mechanisms by which they promote axonal regeneration. Recent findings suggest that the beneficial effect of these cells can be enhanced by genetic engineering approaches or transplanting them with other cell types.Citation103 When compared with Schwann cells and other sources of stem cells, problems of rejection, overgrowth, disease transmission, and ethical issues can be avoided because a subject’s own olfactory mucosa can be used for in vitro studies and clinical trials.Citation106

Challenges in developing cellular therapies for SCI

Cell sourcing

A number of cell sourcing issues need to be considered when developing cell-based therapies for the treatment of SCI and other neurological disorders. To develop such a strategy, a suitable source and quantity of cells for transplantation need to be considered first. If the cells are obtained from an animal source for preclinical studies, there should be a mechanism for translating them into the clinical setting. A second consideration is how to characterize the cells obtained to determine which specific cell phenotype should be implanted for a particular disease. Another issue arises when the stem cells are continuously cultured, including reproducibility of cell lines and the number of times a cell line can be passaged due to variation in passage numbers, feeder layers, and composition of the medium.

Transplantation issues

The first transplantation issue concerns the route of delivery, which should preserve cell integrity and viability. For progenitor cell transplantation, maximum care should be taken to eliminate unwanted differentiation or proliferation, which can lead to formation of teratomas. A second issue is related to the immune response of patients at the time of transplantation, which can lead to graft rejection.

Confirming an experimental therapy before transfer into humans

To obtain the necessary experimental data to begin clinical studies, compelling evidence of benefit must be demonstrated in reproducible animal models of SCI, although no single experimental model exactly mimics clinical conditions in humans. The rodent models that are currently being used include compression, contusion, and transection methods leading to reproducible patterns of structural damage in specific gray and white matter structures. Injury severity varies with each model, so that a spectrum of histopathological and behavioral deficits is reproduced. Also, the SCI patient population is a very heterogeneous group, so no one SCI patient is exactly the same as another. With this in mind, the American Society of Neural Transplantation and Repair comments that the exact type of animal model required will depend on the target condition being considered.Citation105

Cell fate and tracking in vivo

Determination of cell migration and fate in real time and in the long term is of major interest as it relates to dosing, efficacy, optimization, and safety concerns in translational approaches for cell therapy. For both research and clinical purposes, tracking of stem cells after their administration is crucial in vivo and in vitro for determination of their distribution, location, quantity, viability, and final differentiation. There are several strategies to track the cells in vivo and in vitro. Noninvasive strategies include: direct labeling of target cells with paramagnetic contrast agents and tracking them with functional magnetic resonance imaging, using either supermagnetic iron oxide particles,Citation106,Citation107 gadolinium,Citation108,Citation109 19 F isotopes,Citation110 direct labeling with quantum dots using cadmium nanocrystals,Citation111,Citation112 or traditional fluorochromes like PKH26,Citation113 or internal labeling using transfected enhanced green fluorescence protein and firefly luciferase reporter genes via the bioluminescence mechanism.Citation114,Citation115 Each of these cell tracking methods suffers from many drawbacks, including: limited uptake of paramagnetic contrast and weak magnetic resonance imaging signals (except when using the supermagnetic iron oxide particles); traditional fluorochromes being prone to bleaching; cadmium in quantum dot crystals being toxic to cells; and bioluminescence imaging being limited by low tissue penetrance.Citation116

Cell dosing and delivery

In terms of the upper limit and frequency of administration, stem cell dosing remains controversial in the indication of SCI. One would expect an incremental response with dose escalation, but the available data are limited. Recently, Usvald et alCitation117 demonstrated an optimum dosing regimen for intraparenchymal injection of human spinal cord-derived stem cells into the minipig spinal cord for neuronal repopulation, although some studies have suggested that intrathecal and intravenous delivery of stem cells is less efficacious than direct injection into spinal cord tissue.Citation118,Citation119 Vaquero et alCitation118 also showed that intralesional injection of stem cells into rats with SCI produced a better outcome than intravenous administration. A recent study by Wu et alCitation120 proposed fibrin glue as a vehicle for delivering mesenchymal stem cells to injured neural tissues, with good functional recovery, although it is still a challenge to target the stem cells within cellular precision.

Conclusion and future directions

In the field of translational biology, we are rapidly moving toward translation into clinical practice, which provides advanced cell culture techniques and plasticity with regard to choosing different stem cell sources to test the ability of these cells in the treatment of SCI and other neurological disorders. Research using in vitro and in vivo regenerative medicine models has demonstrated the potential use of stem cells in the treatment of various diseases, but uncertainty regarding a suitable source have raised controversial problems concerning their identification and development of new cell-based therapies.

Although various factors are involved in causing different types of SCI, destruction of the signal pathway is the same. Reier et alCitation121 performed the first human fetal stem cell transplantation for SCI and the procedure explicitly recognized the advantage of using stem cells, with a positive outcome in regards to both safety and efficacy. More studies are needed to enhance graft cell viability and application of controlled differentiated cells in SCI. Several other studies have also demonstrated that histological changes in axonal regeneration and sprouting are crucial for behavioral recovery, therefore promoting axonal elongation and sprouting is now a well accepted strategy for treatment of SCI. These cellular-based therapies combined with additional strategies such as neurotrophins, neuroprotective agents, and engineering of neurobiocompatible materials to bridge the cord injury increase the hope of improved treatment of SCI.

Because of the complexity of the issues involved, different viewpoints, and variations between protocols at different institutions for graft replacement transplantation studies, it is difficult to develop a common protocol. Questions regarding relevant animal modeling in both large and small animals also require continued discussion, and controlled preclinical data in Phase I studies are essential to evaluate the safety of cell therapy.

Disclosure

The authors report no conflicts of interest in this work.

References

  • WebbAANganSFowlerJDSpinal cord injury I: A synopsis of the basic scienceCan Vet J20105148549220676289
  • UlzheimerJCPelesELevinsonSRMartiniRAltered expression of ion channel isoforms at the node of Ranvier in P0-deficient myelin mutantsMol Cell Neurosci200425839414962742
  • CaoQLZhangYPHowardRMWaltersWMTsoulfasPWhittemoreSRPluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord are restricted to a glial lineageExp Neurol2001167485811161592
  • CaoQLHowardRMDennisonJBWhittemoreSRDifferentiation of engrafted neuronal-restricted precursor cells is inhibited in the traumatically injured spinal cordExp Neurol200217734935912429182
  • ShihabuddinLSHornerPJRayJGageFHAdult spinal cord stem cells generate neurons after transplantation in the adult dentate gyrusJ Neurosci2000208727873511102479
  • VroemenMAignerLWinklerJWeidnerNAdult neural progenitor cell grafts survive after acute spinal cord injury and integrate along axonal pathwaysEur J Neurosci20031874375112925000
  • HornerPPowerAKempermannGProliferation and differentiation of progenitor cells throughout the intact adult spinal cordJ Neurosci2000202218222810704497
  • ZompaEACainLDEverhartAWMoyerMPHulseboschCETransplant therapy: recovery of function after spinal cord injuryJ Neurotrauma1997144795069300561
  • Di GiovanniSRegeneration following spinal cord injury, from experimental models to humans: where are we?Expert Opin Ther Targets20061036337616706677
  • HerbertsCAKwaMSHermsenHPRisk factors in the development of stem cell therapyJ Transl Med201192921418664
  • ChoppMZhangXHLiYSpinal cord injury in rat: treatment with bone marrow stromal cell transplantationNeuroreport2000113001300511006983
  • TatorCHBiology of neurological recovery and functional restoration after spinal cord injuryNeurosurgery1998426967089574633
  • TatorCHExperimental and clinical studies of the pathophysiology and management of acute spinal cord injuryJ Spinal Cord Med1996192062149237787
  • TatorCHUpdate on the pathophysiology and pathology of acute spinal cord injuryBrain Pathol199554074138974623
  • NorenbergMDSmithJMarcilloAThe pathology of human spinal cord injury: defining the problemsJ Neurotrauma20042142944015115592
  • PangDWilbergerJEJrSpinal cord injury without radiographic abnormalities in childrenJ Neurosurg1982571141297086488
  • WolmanLThe disturbances of circulation in traumatic paraplegia in acute and late stages: a pathological studyParaplegia1965221322614261504
  • AndersonDKHallEDPathophysiology of spinal cord traumaAnn Emerg Med1989229879928503537
  • GeislerFHDorseyFCColemanWPRecovery of motor function after spinal cord injury – a randomized, placebo-controlled trial with GM-1 gangliosideN Engl J Med1991324182918382041549
  • LapchakPAAraujoDMSongDZivinJANeuroprotection by the selective cyclooxygenase-2 inhibitor SC-236 results in improvements in behavioral deficits induced by reversible spinal cord ischemiaStroke2001321220122511340237
  • TatorCHFehlingsMGReview of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanismsJ Neurosurg19917515262045903
  • NashmiRFehlingsMGMechanisms of axonal dysfunction after spinal cord injury: with an emphasis on the role of voltage-gated potassium channelsBrain Res Rev20013816519111750932
  • FrankelHLCollJRCharlifueSWLong-term survival in spinal cord injury: a fifty year investigationSpinal Cord1998362669589527
  • HagenEMLieSARekandTGilhusNEGronningMMortality after traumatic spinal cord injury: 50 years of follow-upJ Neurol Neurosurg Psychiatry20108136819726408
  • BrackenMShepardMHolfordTResults of the third acute spinal cord injury randomized controlled trialJ Neurosurg1998896997069817404
  • BarakatDJGaglaniSMNeravetlaSRSurvival, integration, and axon growth support of glia transplanted into the chronically contused spinal cordCell Transplant20051422524015929557
  • ThompsonPMMousGWCummingsJLTogaAWCortical Variability and Asymmetry in Normal Aging and Alzheimer’s DiseaseCereb Cort199886492509
  • LuJFeronFMackay-SimAWaitePMOlfactory ensheathing cells promote locomotor recovery after delayed transplantation into transected spinal cordBrain2002125142111834589
  • Keyvan-FouladiNRaismanGLiYFunctional repair of the corticospinal tract by delayed transplantation of olfactory ensheathing cells in adult ratsJ Neurosci2003232834
  • VaqueroJZuritaMOyaSSantosMCell therapy using bone marrow stromal cells in chronic paraplegic rats: systemic or local administration?Neurosci Lett200639812913416423458
  • KamadaTKodaMDezawaMTransplantation of bone marrow stromal cell-derived Schwann cells promotes axonal regeneration and functional recovery after complete transection of adult rat spinal cordJ Neuropathol Exp Neurol200564374515715083
  • BiernaskieJSparlingJSLiuJSkin-derived precursors generate myelinating Schwann cells that promote remyelination and functional recovery after contusion spinal cord injuryJ Neurosci2007279545955917804616
  • ErcegSRonaghiMStojkovicMHuman embryonic stem cell differentiation toward regional specific neural precursorsStem Cells200927788718845761
  • LeeHShamyGAElkabetzYDirected differentiation and transplantation of human embryonic stem cell-derived motoneuronsStem Cells2007251931193917478583
  • ErcegSLainezSRonaghiMDifferentiation of human embryonic stem cells to regional specific neural precursors in chemically defined medium conditionsPloS One20083e212218461168
  • LeeGKimHElkabetzYIsolation and directed differentiation of neural crest stem cells derived from human embryonic stem cellsNat Biotechnol2007251468147518037878
  • ErcegSRonaghiMOriaMTransplanted oligodendrocytes and motoneuron progenitors generated from human embryonic stem cells promote locomotor recovery after spinal cord transactionStem Cells2010281541154920665739
  • LiJYChristophersenNSHallVSouletDBrundinPCritical issues of clinical human embryonic stem cell therapy for brain repairTrends Neurosci20083114615318255164
  • ReynoldsBAWeissSGeneration of neurons and astrocytes from isolated cells of the adult mammalian central nervous systemScience1992255170717101553558
  • LuskinMBRestricted proliferation and migration of postnatally generated neurons derived from the forebrain subventricular zoneNeuron1993111731898338665
  • LoisCAlvarez-BuyllaAProliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and gliaProc Natl Acad Sci U S A199390207420778446631
  • SeabergRMVan der KooyDAdult rodent neurogenic regions: the ventricular subependyma contains neural stem cells, but the dentate gyrus contains restricted progenitorsJ Neurosci2002221784179311880507
  • PalmerTDRayJGageFHFGF-2 responsive neuronal progenitors reside in proliferative and quiescent regions of the adult rodent brainMol Cell Neurosci199564744868581317
  • CattaneoEMcKayRProliferation and differentiation of neuronal stem cells regulated by nerve growth factorNature19903477627652172829
  • HsuYCLeeDCChiuIMNeural stem cells, neural progenitors, and neurotrophic factorsCell Transplant20071613315017474295
  • HooshmandMJSontagCJUchidaNTamakiSAndersonAJCummingsBJAnalysis of host-mediated repair mechanisms after human CNS-stem cell transplantation for spinal cord injury: correlation of engraftment with recoveryPloS One20094e587119517014
  • CummingsBJUchidaNTamakiSJHuman neural stem cells differentiate and promote locomotor recovery in spinal cord-injured miceProc Natl Acad Sci U S A2005102140691407416172374
  • Karimi-AbdolrezaeeSEftekharpourEWangJMorsheadCMFehlingsMGDelayed transplantation of adult neural precursor cells promotes remyelination and functional neurological recovery after spinal cord injuryJ Neurosci2006263377338916571744
  • YanJXuLWelshAMExtensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cordPloS Med20074e3917298165
  • BabuHCheungGKettenmannHPalmerTDKempermannGEnriched monolayer precursor cell cultures from micro-dissected adult mouse dentate gyrus yield functional granule cell-like neuronsPloS One20072e38817460755
  • KimBGHwangDHLeeSIKimEJKimSUStem cell-based cell therapy for spinal cord injuryCell Transplant20071635536417658126
  • ZhuJWuXZhangHLAdult neural stem cell therapy: expansion in vitro, tracking in vivo and clinical transplantationCurr Drug Targets200569711015720217
  • CouttsMKeirsteadHSStem cells for the treatment of spinal cord injuryExp Neurol200820936837717950280
  • FitchMTSilverJCNS injury, glial scars, and inflammation: inhibitory extracellular matrices and regeneration failureExp Neurol200820929430117617407
  • AmabileGMeissnerAInduced pluripotent stem cells: current progress and potential for regenerative medicineTrends Mol Med200915596819162546
  • DimosJTRodolfaKTNiakanKKInduced pluripotent stem cells generated from patients with ALS can be differentiated into motor neuronsScience20083211218122118669821
  • WernigMZhaoJPPruszakJNeurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson’s diseaseProc Natl Acad Sci U S A20081055856586118391196
  • KajiKNorrbyKPacaAMileikovskyMMohseniPWoltjenKVirus-free induction of pluripotency and subsequent excision of reprogramming factorsNature200945877177519252477
  • HuangfuDOsafuneKMaehrRInduction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2Nat Biotechnol2008261269127518849973
  • ZhouHWuSJooJYGeneration of induced pluripotent stem cells using recombinant proteinsCell Stem Cell2009438138419398399
  • TakahashiKTanabeKOhnukiMInduction of pluripotent stem cells from adult human fibroblasts by defined factorsCell200713186187218035408
  • YamanakaSA fresh look at iPS cellsCell2009137131719345179
  • AtkinsHFreedmanMImmune ablation followed by autologous hematopoietic stem cell transplantation for the treatment of poor prognosis multiple sclerosisMethods Mol Biol200954923123619378207
  • FreedmanMSBar-OrAAtkinsHLInternational MSCT Study GroupThe therapeutic potential of mesenchymal stem cell transplantation as a treatment for multiple sclerosisMult Scler20101650351020086020
  • OkadaSIshiiKYamaneJIn vivo imaging of engrafted neural stem cells: its application in evaluating the optimal timing of transplantation for spinal cord injuryFASEB J2005191839184116141363
  • NeuhubeBTimothyHBShumskyJSGalloGFischerIAxon growth and recovery of function supported by human bone marrow stromal cells in the injured spinal cord exhibit donor variationsBrain Res20051035738515713279
  • KodaMKamadaTHashimotoMAdenovirus vector-mediated ex vivo gene transfer of brain-derived neurotrophic factor to bome marrow stromal cells promotes axonal regeneration after transplantation in completely transected adult rat spinal cordEur Spine J2007162206221417885772
  • McGuckinCBasfordCHangerKHabibollahSForrazNCord blood revelations: the importance of being a first born girl, big, on time and to a young mother!Early Hum Dev20078373374117942249
  • WattSContrerasMStem cell medicine: umbilical cord blood and its stem cell potentialSemin Fetal Neonatal Med20051020922015927877
  • BallenKBarkerNStewartKGreeneFLaneACollection and preservation of cord blood for personal useBiol Blood Marrow Transplant20081435636318275904
  • PipesBTsangTPengSFiederleinRGrahamMHarrisDTelomere length changes after umbilical cord blood transplantTransfusion2006461038104316734822
  • SlatterMGenneryAUmbilical cord stem cell transplantation for primary immunodeficienciesExpert Opin Biol Ther2006655556516706603
  • LimJParkSOhJBrain-derived neurotrophic factor stimulates the neural differentiation of human umbilical cord blood-derived mesenchymal stem cells and survival of differentiated cells through MAPK/ERK and PI3K/Akt-dependent signaling pathwaysJ Neurosci Res2008862168217818438930
  • KangKKimSOhYA 37-year-old spinal cord-injured female patient transplanted of multipotent stem cells from human UC blood, with improved sensory perception and mobility, both functionally and morphologically: a case studyCytotherapy2005736837316162459
  • DasariVRSpomarDGLiLGujratiMRaoJSDinhDHUmbilical cord blood stem cell mediated downregulation of fas improves functional recovery of rats after spinal cord injuryNeurochem Res20083313414917703359
  • WalczakPChenNEveDLong-term cultured human umbilical cord neural-like cells transplanted into the striatum of NOD SCID miceBrain Res Bull20077415516317683802
  • ChoSRYangMSYimSHNeurally induced umbilical cord blood cells modestly repair injured spinal cordsNeuroreport2008191259126318695504
  • CaplanAIAdult mesenchymal stem cells for tissue engineering versus regenerative medicineJ Cell Physiol2007213234134717620285
  • PittengerMFMackayAMBeckSCMultilineage potential of adult human mesenchymal stem cellsScience1999284541114314710102814
  • CaplanAIMesenchymal stem cellsJ Orthop Res199196416501870029
  • SchäfflerABüchlerCConcise review: adipose tissue-derived stromal cells – basic and clinical implications for novel cell-based therapiesStem Cells20072581882717420225
  • ZukPAZhuMAshjianPHuman adipose tissue is a source of multipotent stem cellsMol Biol Cell2002134279429512475952
  • IzadpanahRTryggCPatelBBiologic properties of mesenchymal stem cells derived from bone marrow and adipose tissueJ Cell Biochem2006991285129716795045
  • RomanovYADarevskayaANMerzlikinaNVBuravkovaLBMesenchymal stem cells from human bone marrow and adipose tissue: isolation, characterization, and differentiation potentialitiesBull Exp Biol Med200514013814316254640
  • ArrigoniELopaSde GirolamoLStancoDBriniATIsolation, characterization and osteogenic differentiation of adipose-derived stem cells: from small to large animal modelsCell Tissue Res2009338340141119882172
  • ZannettinoACPatonSArthurAMultipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivoJ Cell Physiol200821441342117654479
  • ChiGFKimMRKimDWJiangMHSonYSchwann cells differentiated from spheroid-forming cells of rat subcutaneous fat tissue myelinate axons in the spinal cord injuryExp Neurol201022230431720083105
  • ZhangHTChengHYCaiYQComparison of adult neurospheres derived from different origins for treatment of rat spinal cord injuryNeurosci Lett200945811612119394407
  • OhtaYTakenagaMTokuraYMature adipocyte-derived cells, dedifferentiated fat cells (DFAT), promoted functional recovery from spinal cord injury-induced motor dysfunction in ratsCell Transplant20081787788619069631
  • KangSKShinMJJungJSKimYGKimCHAutologous adipose tissue-derived stromal cells for treatment of spinal cord injuryStem Cells Dev20061558359416978061
  • RyuHHLimJHByeonYEFunctional recovery and neural differentiation after transplantation of allogeneic adipose-derived stem cells in a canine model of acute spinal cord injuryJ Vet Sci20091027328419934591
  • OhtaYTakenagaMTokuraYMature adipocyte-derived cells, dedifferentiated fat cells (DFAT), promoted functional recovery from spinal cord injury-induced motor dysfunction in ratsCell Transplant20081787788619069631
  • RaJCShinISKimSHSafety of intravenous infusion of human adipose tissue-derived mesenchymal stem cells in animals and humansStem Cells Dev2011201297130821303266
  • GilsonJMBlakemoreWFSchwann cell remyelination is not replaced by oligodendrocyte remyelination following ethidium bromide induced demyelinationNeuroreport2002131205120812151770
  • BussAPechKKakulasBAGrowth-modulating molecules are associated with invading Schwann cells and not astrocytes in human traumatic spinal cord injuryBrain200713094095317314203
  • FrostickSPYinQKempGJSchwann cells, neurotrophic factors, and peripheral nerve regenerationMicrosurgery1998183974059880154
  • WoodhooASahniVGilsonJSchwann cell precursors: a favourable cell for myelin repair in the central nervous systemBrain20071302175218517550908
  • PapastefanakiFChenJLavdasAAThomaidouDSchachnerMMatsasRGrafts of Schwann cells engineered to express PSA-NCAM promote functional recovery after spinal cord injuryBrain20071302159217417626035
  • GraziadeiPPCLevineRRMontigraziadeiGANeurogenesis and neuron regeneration in the olfactory system of mammals: morphological aspects of differentiation of and structural organization of the olfactory sensory neuronsJ Neurocytol19798118438867
  • FarbmanALOlfactory neurogenesis: genetic or environmental controls?Trends Neurosci1990133623651699323
  • SchwobJNeuronal regeneration and the peripheral olfactory systemAnat Rec2002269334911891623
  • AlexanderCLFitzGeraldUFBarnettSCIdentification of growth factors that promote long-term proliferation of olfactory ensheathing cells and modulate their antigenic phenotypeGlia20023734936411870874
  • RuitenbergMJLevisonDBLeeSVVerhaagenJHarveyARPlantGWNT-3 expression from engineered olfactory ensheathing glia promotes spinal sparing and regenerationBrain200512883985315716305
  • FéronFPerryCCochraneJAutologous olfactory ensheathing cell transplantation in human spinal cord injuryBrain20051282951296016219671
  • RedmondDEJrFreemanTCommentary: The American Society for Neural Transplantation and Repair Considerations and Guidelines for Studies of Human Subjects. The Practice Committee of the Society. Approved by CouncilCell Transpl200110661664
  • SykovaEJendelovaPIn vivo tracking of stem cells in brain and spinal cord injuryProg Brain Res200716136738317618991
  • KallurTFarrTDBohm-SturmPKokaiaZHoehnMSpatio-temporal dynamics, differentiation and viability of human neural stem cells after implantation into neonatal rat brainEur J Neurosci20113438239321707793
  • RudeliusMDaldrup-LinkHEHeinzmannUHighly efficient paramagnetic labelling of embryonic and neuronal stem cellsEur J Nucl Med Mol Imaging2003301038104412567250
  • LiuYHeZJXuBEvaluation of cell tracking effects for transplanted mesenchymal stem cells with jetPEI/Gd-DTPA complexes in animal models of hemorrhagic spinal cord injuryBrain Res20111391243521420939
  • Boehm-SturmPMenglerLWeckerSHoehnMKallurTIn vivo tracking of human neural stem cells with 19F magnetic resonance imagingPLoS One20116e2904022216163
  • LuSXuXZhaoWTargeting of embryonic stem cells by peptide-conjugated quantum dotsPLoS One20105e1207520711469
  • ShahBSMaoJJLabeling of mesenchymal stem cells with bioconjugated quantum dotsMethods Mol Biol2011680617521153373
  • Seyed JafariSSAli AghaeiAAsadi-ShekaariMNematollahi-MahaniSNSheibaniVInvestigating the effects of adult neural stem cell transplantation by lumbar puncture in transient cerebral ischemiaNeurosci Lett20114951521333715
  • DaadiMMLiZAracAMolecular and magnetic resonance imaging of human embryonic stem cell-derived neural stem cell grafts in ischemic rat brainMol Ther2009171282129119436269
  • SwijnenburgRJSchrepferSCaoFIn vivo imaging of embryonic stem cells reveals patterns of survival and immune rejection following transplantationStem Cells Dev2008171023102918491958
  • De AlmeidaPEvan RappardJRWuJCIn vivo bioluminescence for tracking cell fate and functionAm J Physiol Heart Circ Physiol2011301H663H67121666118
  • UsvaldDVodickaPHlucilovaJAnalysis of dosing regimen and reproducibility of intraspinal grafting of human spinal stem cells in immunosuppressed minipigsCell Transplant2010191103112220412634
  • VaqueroJZuritaMOyaSSantosMCell therapy using bone marrow stromal cells in chronic paraplegic rats: systemic or local administration?Neurosci Lett200639812913416423458
  • KhalatbaryARTiraihiTA comparative study of therapeutic benefits of intraspinal and intravenous bone marrow stromal cell administration to spinal cord injuriesIran Biomed J200913434819252677
  • WuXRenJLiJFibrin glue as the cell-delivery vehicle for mesenchymal stromal cells in regenerative medicineCytotherapy20121455556222175911
  • ReierPJCellular transplantation strategies for spinal cord injury and translational neurobiologyNeuro Rx2004142445115717046