Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81 Warning: fopen(/home/virtual/e-kjs/journal/upload/ip_log/ip_log_2024-04.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84 Advances in Neural Stem Cell Therapy for Spinal Cord Injury: Safety, Efficacy, and Future Perspectives
Neurospine Search

CLOSE


Neurospine > Volume 19(4); 2022 > Article
Lee, Nam, Joo, and Lee: Advances in Neural Stem Cell Therapy for Spinal Cord Injury: Safety, Efficacy, and Future Perspectives

Abstract

Spinal cord injury (SCI) is a devastating central nervous system injury that leads to severe disabilities in motor and sensory functions, causing significant deterioration in patients’ quality of life. Owing to the complexity of SCI pathophysiology, there has been no effective treatment for reversing neural tissue damage and recovering neurological functions. Several novel therapies targeting different stages of pathophysiological mechanisms of SCI have been developed. Among these, treatments using stem cells have great potential for the regeneration of damaged neural tissues. In this review, we have summarized recent preclinical and clinical studies focusing on neural stem cells (NSCs). NSCs are multipotent cells with specific differentiation capabilities for neural lineage. Several preclinical studies have demonstrated the regenerative effects of transplanted NSCs in SCI animal models through both paracrine effects and direct neuronal differentiation, restoring synaptic connectivity and neural networks. Based on the positive results of several preclinical studies, phase I and II clinical trials using NSCs have been performed. Despite several hurdles and issues that need to be addressed in the clinical use of NSCs in patients with SCI, gradual progress in the technical development and therapeutic efficacy of NSCs treatments has enhanced the prospects for cell-based treatments in SCI.

INTRODUCTION

Traumatic spinal cord injury (SCI) is a catastrophic event with a high mortality rate and causes physical and emotional difficulties in patients.1-5 It is defined as injury to the spinal cord, nerve roots, osseous structures, and disco-ligamentous components [6]. The subsequent formation of reactive tissue scarring and cystic cavitation results in the development of molecular and physical barriers to regenerative axonal growth and long-term neurological deficits in SCI. The prevalence and incidence of SCI vary according to geopolitical and economic conditions, and approximately 1,000 new cord injury cases occur every year in South Korea [7]. Although the global incidence is similar between sexes, men have a higher incidence than women aged 20–40 years [8]. Moreover, as the global population tends to grow and health care systems improve, an increase in the absolute number of people living with SCI is expected [1,3,4].
Anti-inflammatory methylprednisolone sodium succinate is the first-line drug treatment for patients with SCI [9]. After initial management, clinicians surgically decompress the spinal cord and control the lesion site if needed [10]. Many studies have been conducted to prevent or reduce the effects of secondary injury; among them, research on steroids and neuroprotective alternatives has been discussed for a long time. For the regeneration of damaged neural cells in SCI, various types of stem cells including Schwann cells, olfactory ensheathing cells, embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), and neural stem cells (NSCs), have been examined preclinically and in animal models of SCI.
Recently, transplantation of NSCs has been shown to promote the repair or regeneration of damaged spinal cords. In this review article, we have discussed the characteristics, origin, and recent developments of NSCs in clinical trials of SCI.

BASIC CHARACTERISTIC OF STEM CELLS

Stem cells exhibit 2 characteristics: self-renewal and multipotency. ESCs that are established from fertilized eggs can satisfy the definition of stem cells because ESCs can proliferate indefinitely and differentiate into whole body [11]. Recently, induced pluripotent stem cells (iPSCs) have been suggested to exhibit characteristics similar to those of ESCs [12]. These 2 types of stem cells are called pluripotent stem cells (PSCs). In contrast, adult stem cells (ASCs) reside in organs and regenerate their tissues when damaged [13]. Therefore, ASCs usually have limited lifespan and differentiation potential. In clinical trials to regenerate the damaged central nervous system (CNS), 2 types of ASCs have been used: MSCs and NSCs. The key feature of MSCs is their differentiation potential into mesodermal tissues, such as osteoblasts, adipocytes, chondrocytes, and even other lineages [14]. Moreover, MSCs produce various paracrine factors that have beneficial effects on regeneration and immune modulation [15]. However, several studies have concluded that their beneficial effects are due to functional modulation, and not by direct neuronal regeneration and integration into the injured CNS [16,17]. NSCs are characterized by the expression of typical markers, such as Nestin or Sox2 [18]. Generally, they reside in the subventricular zone and the subgranular zone [19,20], which are specialized niches where young neurons for the olfactory bulb and hippocampus, respectively, are generated [21]. NSCs can self-renew and play a role in neurogenesis in the adult brain [22,23]. NSCs preferentially differentiate into neural lineages such as neurons, astrocytes, and oligodendrocytes, which are attractive for clinical use in CNS diseases [24]. NSCs also secrete beneficial paracrine factors that can help regenerate the damaged CNSs [25-27]. Such characters make NSCs a potent and versatile cellular drug candidate for the treatment of the CNS injuries.

ESTABLISHMENT OF NSCs

NSCs have been established from several sources [28]. Among them, the conventional source of NSCs is the fetal CNS [8,26,29]. Fetal NSCs (fNSCs) have self-renewal potential and neural differentiation capacity [30]. The therapeutic potential of fNSCs has been demonstrated in a model of SCI [8,29,31], and interestingly, human fNSCs showed neurogenesis after injection into immunodeficient mice in vivo [8,32]. fNSCs can differentiate into neurons, which can connect with surrounding neurons [8,29,31]. With promising data from several preclinical studies, most clinical trials have used NSCs derived from the human fetal CNS, including the brain and spinal cord [33]. However, unavoidable ethical issues using fetal CNS are critical for commercial development, and they provide a strong motivation for other cellular sources.
One candidate is the adult NSCs (aNSCs), which can be isolated from the adult CNS. The adult olfactory bulb is the source of NSCs. The olfactory bulb core is an extension of the rostral migratory stream and is thus a potential source of neural progenitors and NSCs [34]. Human spine is another option for aNSCs transplantation [35-37]. Through several preclinical studies, the beneficial effects of aNSCs have been proven in SCI models [32,38-43]. These studies suggested that the beneficial effects of aNSCs come not only from their paracrine effects in neural tissue repair and regeneration, but also from their direct differentiation into various neuronal lineage cells that are integrated and form neuronal networks with the host CNS. This multiple recovery mechanism implies that aNSCs could be an optimal choice in the treatment of SCI.
Despite these advantages, technical difficulties remain to be solved in order to utilize these cells in real-world clinical practice. For the appropriate use of aNSCs, they must be properly isolated and effectively increased in number. Compared with other stem cells, aNSCs reside in relatively restricted areas of the adult CNS [44]. In addition, they have limited and different proliferation capacities according to the lesion type and location [45]. To address the technical difficulties in primary isolation and stable in vitro expansion of aNSCs, several research teams have suggested various scientific and technical approaches [46]. Surgical samples from adult CNS are usually very small (1–2 mL) and the number of resident aNSCs within the tissue is also very small. Therefore, aNSCs isolation techniques have been optimized to increase the success rate of primary isolation. First, CNS tissues were physically minced and enzymatically digested into single cells. Enzymatic digestion is a critical step because it directly affects NSC survival. Papain, trypsin, and collagenase have been commonly used, and in some reports, papain dissociation was suggested to be optimal for the primary isolation of aNSCs [47,48]. After mechanical and enzymatic dissociation of CNS tissues, isolated single cells expand in number. There are 2 alternative culture methods in use: the neurosphere and adherent culture methods. Conventionally, the neurosphere culture method has been used for in vitro culture of NSCs [20,49-57]. However, difficulties in stable in vitro expansion of aNSCs using suspension culture methods require the development of other culture methods. Moreover, a single neurosphere may not be derived from a single NSC [58]. The possible heterogenic origin of neurospheres could not guarantee the homogeneity of in vitro-expanded aNSCs in suspension culture conditions [59-61]. To overcome the limitations of the neurosphere culture method, an alternative adherent culture methods for NSCs, was developed [52,62-64]. In this method, each group used its coating plates to attach NSCs to the plates and various culture medium compositions. Laminin and poly-L-ornithine (PLO) have been used to coat plate frequently, which increase the adherent efficiency of NSCs. To maintain stemness and proliferation of NSCs, the amount of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) have been optimized [65]. For example, we expanded aNSCs from temporal lobectomy samples of epilepsy patients without any neoplastic diseases on PLO-coated dishes in a DMEM/F12 media supplemented with 1% B27, 1% penicillin/streptomycin cocktail, EGF (50 ng/mL), bFGF (50 ng/mL), and 0.5% fetal bovine serum [62]. Using the adherent culture method, aNSCs were expanded in vitro from 104 to 1012 cells within 8 subcultures for 2 months. Moreover, expression of NSC markers such as nestin and SOX2 maintained stably [62]. If the number of aNSCs required for transplantation is 107 per patient, at least one hundred thousand patients could be treated with a primary culture of aNSCs.
Recently, technical developments have resulted in the establishment of NSCs from ESCs or iPSCs [26]. When ESCs and iPSCs are induced to differentiate into NSCs by several inducers, such as growth factors and cytokines [34], these NSCs have similar characteristics to fNSCs, which can induce neurogenesis in the CNS of immunodeficient mice [66,67]. In several preclinical studies, the therapeutic potential of NSCs derived from ESCs or iPSCs has been demonstrated in animal models of SCI [8,26,29,31]. To date, human clinical data using ESCs or iPSCs for SCI treatment are scarce. Only 2 clinical trials (one in each ESCs and iPSCs) are ongoing right now. Compared to the other cellular sources, iPSCs have great advantages in ethical issues and immune rejection [8]. Therefore, interests in NSCs from iPSCs will continuously increase with the advances with iPSCs technology.

PRECLINICAL STUDY OF NSC FOR SCI

Preclinical studies should be designed to address the activity and safety of stem cell-based products for clinical use. Information about the potential mechanism of action of stem cells in the disease indication, the timing of intervention with respect to disease course, and the mode of delivery to the site of action must be investigated in preclinical models. Many preclinical studies using NSCs in animal models of SCI have been reported in the literature [8,26,29,31], and the therapeutic potential, safety, and several technical aspects of NSCs transplantation have been tested under various conditions.
The characteristics of experimental studies using NSCs are summarized in Table 1. NSCs treatments have been tested at various stages of SCI: acute, subacute and chronic. Mice and rats are the most used animals. In a few studies, human NSCs have been tested in non-human primates [68-70]. The thoracic spinal cord has been the most frequently studied region, where the injury is made by mechanical trauma, such as dropping weight or clip compression. As sudden contusive injury to the cervical spinal cord can be life-threatening, hemitransection is the preferred method for models of cervical SCI. Functional testing scales are somewhat standardized according to animal model species. In mice and rats, the Basso mouse scale or Basso-Beattie-Bresnahan test and CatWalk gait analysis are the most frequently used scales. In addition, many studies have used the von Frey test to evaluate the sensory function. Several studies have also reported functional recovery after transplantation of NSCs as well as graft survival, differentiation and axonal regeneration [40,42,43,68-81].
Although a number of studies have reported promising results of NSCs treatment in SCI, we still have a long way to go to use NSCs in real-world clinical practice. To move from bench to bedside, determining the differences between the animal model and human SCI and closing the gap caused by inherent limitations of the model should be the first step. In general, there are no reliable animal models that can predict human diseases. Under such circumstances, using a model that most closely represents the critical features of the intended indication is the best alternative. Most human SCI cases are caused by mechanical injury. Consequently, we have developed several animal models of SCI using mechanical trauma to the spinal cord. However, the regenerative potential and physical size of the spinal cord differs among species. Considering that our knowledge regarding the pathophysiological mechanism of SCI is limited, there is a need for multiple animal models to properly address the delivery, efficacy, toxicity and tumorigenicity of NSCs in SCI treatments.

STRATEGIES FOR CLINICAL TRANSITION OF NSCs

In addition to the general consideration of clinical transition from preclinical studies, more knowledge of NSCs needs to be elucidated. The key questions that remain unanswered are as follows: (1) What is the optimal timing for treatment? (2) What are the optimal combinatory or supplementary measures for successful treatment? (3) What is the optimal route of administration? (4) How many cells should be transplanted? (5) Which cellular source should be used, with regard to efficacy, utility, and safety?

1. Optimal Timing for NSC Treatment

Since glial scarring is one of the major barriers to axonal growth and reintegration into neural circuits at the lesion site, cell grafting in the acute phase of SCI might be more beneficial than treatment in chronic-phase SCI. Cheng et al. [82] tested 3 different timings of human NSCs injection (acute, subacute, and chronic) in a T10 contusion injury rat model. The subacute group showed more prominent functional improvements than the chronic group, which supports the idea of the early treatment of SCIs. Furthermore, several studies have suggested that NSCs exert beneficial effects by suppressing neuroinflammation [27,40]. These findings imply that NSC transplantation may benefit the acute to subacute phase of SCI, the period during which the most active inflammatory process takes place. However, several studies have reported contradictory findings. Nguyen et al. [83] injected human NSCs into mice immediately after T9 contusive SCI, and the donor cells showed astroglial differentiation near the lesion but failed to produce functional improvements. In contrast, Salazar et al. [43] transplanted human NSCs into mice 1 month after T9 spinal cord contusive injury, and NSC transplanted mice demonstrated significantly improved locomotor recovery. Therefore, it is difficult to determine that which time window would be the most beneficial for transplanting NSCs after SCI, and we need more data for validation. Many preclinical studies have shown that grafted NSCs survive, migrate and integrate into the injured spinal cord, and differentiate into 3 CNS cell lineages. This suggests that data is insufficient to set specific time window for successful NSCs treatment, and more studies are required to verify the effective treatment timing for NSCs therapy.

2. Considered Combinatory or Supplementary Measures for Successful NSC Treatment

Since SCI is a complicated process with multiphasic cellular and molecular responses that vary over time [8], testing various strategies in patients with different injury time windows and situations is important along with efforts to find the best time window for the treatment of SCI patients. It is clear that a single treatment modality is not effective in SCI treatment. Several combinatory treatments to enhance grafted cell survival, migration, differentiation and axonal regeneration along with functional recovery have been studied. Synergic treatments with neurotrophic factors such as EGF, bFGF, platelet-derived growth factor, and neurotrophin-3 by implanting genes in NSCs [41,76]. In addition, mixing other cells such as fibroblasts or neuroepithelial-like stem cells with transplanted NSCs to enhance structural repair [84,85]; cotreatment with growth factor cocktail [42,77,86,87]; and adding rehabilitation exercise [81] also showed promising results. In chronic-phase SCI, pretreatment with chondrotinase ABC (ChABC) before transplantation of NSCs seemed to unlock scar tissue around the injury sites and produce a microenvironment conducive for NSCs regeneration. Several preclinical studies that have tested the efficacy of ChABC pretreatment in chronic SCI have also shown locomotor improvements [41,88-90]. These studies are in progress. In the future, there is a need to develop a comprehensive protocol by combining effective strategies according to the injury timeline.
One of the most promising combinatory treatments for NSCs is the use of tissue-engineered scaffolds. The use of scaffolds may act as a bridge that fills the lesion gap and helps reconnect and recover neural networks. Several scaffold types have been developed and tested in preclinical studies. Günther et al. [91] reported that anisotropic alginate hydrogel scaffolds promote axonal regrowth and guided regenerated axons. Huang et al. [92] had used similar scaffolds and demonstrated axonal regrowth through these scaffolds in chronic SCI after the lesion scar was removed. In addition, they have shown significant improvements in functional outcomes and electrophysiological conductivity. Nguyen et al. [83] reported 3-dimensional aligned nanofiber-hydrogel scaffolds could be effective. Furthermore, several other types of scaffolds, such as taxol-modified collagen scaffolds [93], graphene oxide scaffolds [94], nanostructured composite scaffolds [95], have shown efficacy in axonal regeneration. Several studies have tested combinatory treatment with NSCs, and several types of scaffolds have reported reductions in lesion cavities, enhanced grafted cell survival and axonal regeneration, and functional improvements [42,69]. However, the results have not been consistent in other studies [72,96]. Clearly, various types of scaffolds have shown their efficacy in providing anatomical, structural, and histological framework which can guide and promote axonal regeneration. These scaffolds can replace the injured tissue gap, which would not be possible for regenerating axons to pass through and may help the axons to overpass the lesion site. Future studies are required to verify the role of scaffolds in combination with stem cell-based treatments for SCI.

3. Administration Routes

The issue of NSCs administration routes is also a complicated question that needs to be addressed. Three injection routes are possible for SCI treatment and have been tested: intrathecal, intraspinal, and intravenous. As shown in Table 1, most preclinical studies using NSCs used the intraspinal route for cell transplantation. Amemori et al. [97] compared the intrathecal and intraspinal administration routes in an acute contusive SCI model. Both the methods facilitated functional locomotor recovery; however, cell graft survival at the lesion stie was better in the intraspinal injection group, and they concluded that intraspinal transplantation would be more helpful for long-term spinal cord tissue regeneration. Nevertheless, evidence favoring intrathecal injection as an administration route is also available. Cheng et al. [98] transplanted human NSCs into a contusive rat model of SCI both locally and distally, and significant functional recovery was observed in the distally injected group. Most researchers agree that these beneficial effects arise mainly from the paracrine effects of NSCs. Although these administration routes are clearly disadvantageous in terms of direct neuronal differentiation and tissue regeneration, the intrathecal or intravenous route is a more minimally invasive approach than intraspinal injection, and it can be performed much easier in real-world clinical settings, especially for treating patients in the acute stage of SCI. In summary, the most effective administration route for NSCs transplantation seems to be intraspinal injection. More studies to standardize intraspinal injection procedure and verify its efficacy. In addition, there is also a need for seeking the potential utility of intrathecal or intravenous cell injection in SCI treatment.

4. Number of Cells Needed for Transplantation

The number of cells that should be transplanted to obtain a positive result is another unanswered question. Preclinical studies typically provide the basis for determining the starting human dose. The dose of stem cells is dependent on their stability because effective number of stem cells should be maintained before administration. The number of transplanted cells in preclinical studies presented in the literature ranged between 1× 105 and 4× 107 cells per kilogram of animal body weight [99]. Referring to Table 1, most preclinical studies NSCs have used approximately 5× 105 to 1× 106 cells for intraspinal cell transplantation. Yousefifard et al. [99] suggested that higher cell doses (> 3 × 106 cells/kg) are optimal for transplantation. However, a few studies suggest that there is a certain threshold for the number of transplanted stem cells to survive, and there is no correlation between the number of transplanted cells and functional recovery [26]. Further studies are needed to determine the optimal range of transplanted cell numbers, not only in animal models but also in humans.

5. Issues in Cellular Sources of NSCs

Finally, the cellular source that should be used to obtain NSCs is also an important question in stem cell treatment in SCI. Various cellular sources have been tested. Graft survival, neuronal differentiation and functional recovery have been demonstrated in most preclinical studies where allogeneic NSCs from the fetal brain and spinal cord, as well as human NSCs were transplanted in mouse and rat models [40,75,77,78,81,86,87,100-102]. So far, it seems no specific NSC line showed significant comparative advantage over others. This means that all of NSCs from different cellular sources and lineages should be explored further for their efficacy and safety.
Tumorigenicity and immune rejection are the 2 most important concerns regarding cellular sources. In terms of tumorigenicity, there are numerous experimental design parameters to consider, including the choice of animal model, study duration, route of administration, number of cells tested, positive control selection, and the definition of a positive result. The selected animal model should allow sufficient survival of the stem cell product to enable the assessment of potential tumorigenicity. Therefore, immunocompromised rodents are frequently used for this purpose. Likewise, the study duration should be sufficient to permit the detection of potential tumors. Tumorigenicity studies lasting 9–12 months have been requested by regulatory agencies. To date, reports of tumor formation in NSCs treatments in animal models of SCI are scarce. However, Salewski et al. [80] reported that primitive NSCs derived from ESCs could be transformed into teratomas. Tumorigenicity potential may also reside in NSC lineages and should be closely monitored in future studies.
Ready-made NSCs, which are usually obtained from allogeneic brains or spinal cords, have been used in most preclinical studies [40,41,72,74-78,102-104]. This may be due to limitations in time and autologous source tissue to obtain a sufficient number of NSCs for transplantation. Human NSCs were tested in several animal models with promising results [39,70,71,77,79,80,83,86,98,101]. Long-term survival of grafted cells is necessary for locomotor functional recovery [32]. For graft survival, either immunosuppressants were administered after transplantation or nude mice and nude rats were used. Such conditioning for experimental purposes is possible at a preclinical level. However, the use of immunosuppressants in human patients with acute or subacute stage SCI might be risky. SCIs are usually combined with severe multiple traumatic injuries affecting multiple organs and musculoskeletal regions and using immunosuppressants in such conditions poses high risk for sepsis. Transplanted NSCs have paracrine effects which help SCI recovery, even in the absence of graft survival [25]. Nonetheless, considering that grafted cell survival with neuronal differentiation and integration into the host neuronal network would be a favorable long-term outcome, issues regarding immune rejection should be thoroughly studied. The immune rejection issue has brought iPSCs into the spotlight. With the advantage of avoiding ethical issues, autologous iPSCs have become one of the most attractive cell sources for human NSCs. However, a vast number of studies are required to ensure the efficacy, feasibility, and safety of iPSCs in SCI treatment.

CLINICAL TRIALS USING NSCs

In contrast to the relative abundance of preclinical studies on NSCs transplantation in animal models of SCI, clinical trials of NSCs treatment in patients with SCI, which have been published in the literature have been scarce (Table 2). It is encouraging that several studies reported its procedural safety as well as partial success in functional recovery after NSCs transplantation in patients with SCI [105-108]. However, since the number of enrolled patients was small, and only patients in the subacute (within 1 week to 6 months from the injury) and chronic (over 6 months from the injury) phases of SCI were included in most trials, it is quite difficult to conclude therapeutic efficacy of NSCs, especially in acute phase SCI.
The paucity of clinical trials implies difficulty in translation from bench to bedside in SCI research. The fundamental limitation of translational research is the anatomical difference between experimental animal models and humans. In addition, stem cell therapy in animal models had shown inconsistent results regarding functional recovery. The therapeutic potential of NSCs in SCI treatment was observed, but the absence of a certain, reliable modality resulted in numerous exploratory studies that were far from standardization. Consequently, practical questions such as the location and route of cell transplantation, adequate number of transplanted cells, assessment tools and protocols and variability in NSCs generation are still unknown. Furthermore, real-world problems, such as setting a reliable and safe logistic to obtain, store and deliver NSCs for clinical use, recruiting patients, and running clinical trials, would be expensive. Several ongoing clinical trials have been attempted despite of hurdles mentioned above [8,29]. However, extensive efforts to find major breakthroughs in SCI treatment are still needed.

CONCLUSION

NSCs are self-renewing and multipotent stem cells that can differentiate into neural lineage cells. For the past 2 decades, many preclinical studies have tested efficacy and safety of NSCs in several animal models of SCI. Successful neuronal differentiation, replacing damaged neural tissue, and functional improvement were observed in several studies. In addition, NSCs secrete neurotropic factors that help protect or regenerate injured spinal cord. In preclinical level, transplantation of NSCs has been proved as a promising therapeutic approach for SCI treatment. However, some of clinical trials of NSCs did not show enough efficacy as expected. These results suggest that a need for further assessment, and the exact mechanism by which NSCs transplantation improves outcomes after SCI should be explored further.
For future perspective, further data such as treatment benefits in terms of neuronal regeneration and functional recovery, adjustments in dose and administration period, optimal injection route, safety, and the most promising cell source for obtaining NSCs should be acquired and verified through future studies. Moreover, matching preclinical animal models and human SCI is another major hurdle to overcome. Finally, it is also important to highlight that a single treatment modality alone may not be sufficient to treat SCI. In addition to cellular transplantation, combinatory therapies such as neurotrophic and growth factors, the use of scaffolds, and neurorehabilitation may be necessary. Their optimal combination and efficacy should also be verified in future studies. Despite these uncertainties, numerous preclinical studies and clinical trials have reported promising results with NSCs treatment for SCI. We are convinced that NSCs have a potential to make a major breakthrough in SCI treatment in the near future.

NOTES

Conflict of Interest

The authors have nothing to disclose.

Funding/Support

This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author Contribution

Conceptualization: HN, KJ, SL; Data curation: SL, HN; Formal analysis: SL; Methodology: SL, HN, KJ, SL; Project administration: SL; Visualization: SL, HN; Writing - original draft: SL, HN, SL; Writing - review & editing: SL, KJ, SL.

Table 1.
Summary of preclinical studies using neural stem cells/neural progenitor cells in animal spinal cord injury models in literature
Study Species Injury location SCI model Transplantation time after SCI Cell type Cell source Route and dose Combination Functional evaluation Result
Pfeifer et al. [84] 2006 Rat Cervical Transection 8 Weeks Auto/allogenic NPCs Rat brain Intralesional injection, 2.4× 105 cells Fibroblasts N/A Showed substantial axonal regeneration
Nomura et al. [104] 2008 Rat Thoracic Transection 0 Day NSCs Rat brain, spinal cord Intralesional grafts, N/A for cell dose Chitosan channel BBB test Astrocytic, oligodendrocytic differentiation observed in the channels
No functional improvements
Olson et al. [96] 2009 Rat Thoracic Transection 0 Day NSCs Rat brain Intralesional grafts, 4.76 × 105 cells PLGA polymer scaffold BBB test Facilitated axonal regeneration
No functional recovery
Bozkurt et al. [103] 2010 Rat Thoracic Clip compression injury 3 Weeks NSCs SC of transgenic rats Intralesional, 1 × 106 cells Chitosan channel BBB test No functional improvements
Karimi-Abdolrez et al. [41] 2010 Rat Thoracic Clip compression injury 6 Weeks NSCs/NPCs Mouse fetal brain Intralesional, 4 × 105 cells ISD BBB test Promoted axonal integrity, plasticity of the corticospinal tract
Chondrotinase ABC, EGF, bFGF, PDGFAA Grid walking test
Von Frey test Enhanced the plasticity of descending serotonergic pathways
Kusano et al. [76] 2010 Rat Thoracic Clip compression injury 6 Weeks NPCs Rat fetal brain Perilesional injections (4 points around lesion cavity), 2.5 × 105 cells, each NT-3 BBB test Enhanced myelin formation
Partial improvements of hindlimb motor
Pritchard et al. [69] 2010 Monkey Thoracic Hemisection 0 Day Human NSCs N/A Intralesional grafts, 1 × 106 cells PLGA polymer scaffold Ambulation chamber video observational neuromotor score Improvements in postures and movements of leg, foot and toe
ISD
Salazar et al. [43] 2010 Mouse Thoracic Drop weight Contusion injury 1 Month Human NSCs Human fetal brain Perilesional injections, 5 × cells None BMS score, Cat-Walk test, von Frey test Differentiation into oligodentrocytes and neurons as well as astrocytes
Showed locomotor recovery
Yamane et al. [70] 2010 Monkey Cervical Drop weight Contusion injury 9 Days Human NSCs Human fetal brain Perilesional injection, 1 × 106 cells ISD Spontaneous movements, Bar grip strength, treadmill test Motor function improvements
Galectin-1
Du et al. [72] 2011 Rat Thoracic Transection 0 Day NSCs Hippocampus of rat pups Cord lesion site, cell dose not specified PLGA scaffold BBB test Transfected NSCs, co-cultured with scaffold showed the smallest tissue defects at the injury site.
LacZ, NT-3, TrkC gene modification Inclined-grid climbing test Functional improvements observed.
Limited ability of corticospinal tract axonal regeneration
Cheng et al. [71] 2012 Rat Thoracic Drop weight contusion injury 0 Day Human NSCs Human fetal NSCs Either intrathecal or perilesional SC lesion, 5 × 105 cells None BBB test Functional improvements in both intrathecal and perilesional injections
Lu et al. [42] 2012 Rat Cervical Hemisection 2 Weeks Rat and human NSCs Rat fetal SC Intralesional grafts, N/A for cell dose Fibrin matrices with growth factor cocktail BBB test Grafted cells differentiated into multiple cellular phenotypes.
Thoracic Transection Human fetal SC Long axon growths with abundant synapses with hos cells
Improved motor functions
Amemori et al. [39] 2013 Rat Thoracic Balloon-induced compression injury 1 Week Human NSCs Human fetal spinal cord Intralesional, 5 × 105 cells ISD BBB, Plantar, walking-beam test Significant motor, sensory function recovery
Showed robust cell survival and partial lesion filling
Kumamaru et al. [102] 2013 Mouse Thoracic Drop weight contusion injury 12 Weeks NSCs/NPCs Mouse fetal brain Perilesional injections (both rostral and caudal side), 5 × 105 cells, each None BMS score No improvement in motor function
Grip walk test
Footprint analysis
Nemati et al. [68] 2014 Monkey Thoracic Drop weight contusion injury 10 Days NSCs Monkey brain Intralesional injection, 1 × 106 cells/kg None Tarlov scale and tail movements In all scales, transplanted group was faster in recovery.
Limb and tail pinch test
Salewski et al. [80] 2015 Mouse Thoracic Clip compression injury 1 Week NSCs Murine embryonal stem cell Perilesional injections, 5 × 104 cells ISD BMS score, Cat-Walk test, von Frey test Differentiation to oligodendrocytes
Promote remyelination and axonal function
Motor function improvements
Cheng et al. [40] 2016 Mouse N/A N/A 1 Week NSCs Mouse fetal brain N/A None BMS score Improvements in BMS scores
NSC transplantation may modulate SCI-induced inflammatory responses.
Cheng et al. [98] 2016 Rat Thoracic Drop weight contusion injury 4 Weeks Human NSCs Human fetal NSCs Either intrathecal or perilesional SC lesion, 5 × 105 cells None BBB test Functional improvement in intrathecal group
No functional improvements in perilesional injection group
Jin et al. [74] 2016 Rat Thoracic Drop weight contusion injury 13 Weeks NPCs SC of transgenic rats Intralesional & rostral and caudal perilesional injections, 1 × 105 cells each ISD BBB test Similar functional improvements between the treatment groups
Chondrotinase Neurotrophic factors Grid test
Von Frey test Rats treated with NPC with chondrotinase and neurotrophins showed the most significant improvements in bladder function.
Bladder function test
Kadoya et al. [75] 2016 Rat Thoracic Transection 2 Weeks NPCs SC of rat and mouse Intralesional, 1–2 × 106 cells None Staircase task Cell graft survived
Mouse Cervical Positive axonal corticospinal tract regeneration and functional synaptic formation
Improved forelimb function
Tashiro et al. [81] 2016 Mouse Thoracic Drop weight Contusion injury 7 Weeks NSCs/NPCs Mouse fetal brain Intralesional injection, 5 × 105 cells Treadmill training BMS score, von Frey test, Hargreeves plantar test Improved motor and sensory functions
Lu et al. [77] 2017 Rat Cervical Hemisection 2 Weeks Human NSCs Human ESCs Perilesional injections (6 points around lesion cavity), 2.5 × 105 cells, each Growth factor cocktail Forepaw placements on gridwalk task More than a year later, forelimb motor function improved and astrocytes migrated to host tissue.
Nguyen et al. [83] 2017 Mouse Thoracic Drop weight contusion injury 0 Day Human NSCs Human fetal brain Perilesional injections, 1.875 × 104 cells Anti-Ly6G CatWalk behavioral test Showed astroglial differentiation
IgG2a No locomotor improvements
Robinson et al. [87] 2017 Rat Cervical Hemisection 2 Weeks NPCs Rat spinal cord Intralesional injection, 6.25 × 105 cells 4-growth factor cocktail N/A Enhanced graft survival, neuronal differentiation
Thoracic Drop weight Contusion injury Reduction of the lesion sites
Hosseini et al. [73] 2018 Rat Thoracic Clip compression injury 3 Days MSCs/NSCs Rat bone marrow/rat fetal brain Perilesional injection (both rostral and caudal side) MSCs BBB test Most functional improvement in MSCs/NSCs combined treatment group
Nori et al. [89] 2018 Rat Thoracic Clip compression injury 7 Weeks Human NPCs Human bone marrow somatic cells Intralesional injection, 4 × 105 cells Chondrotinase ABC BBB test, Cat-Walk behavioral test, von Frey test Enhanced NPC survival, migration and oligodendrogenic differentiation
Promoted synapse preservation, and enhanced myelination of axons
Showed functional improvements
Riemann et al. [78] 2018 Rat Cervical Clip compression injury 10 Days NPCs Rat fetal brain Perilesional injection, 4 points 1 × 105 cells each None BBB test, Cat-Walk test, Grid walk test Showed differentiation along the oligodendroglial lineage and longterm survival
Reduction in inflammatory cells and markers, apoptosis
Showed functional improvements
Rosenzweig et al. [79] 2018 Monkey Cervical Hemisection 2 Weeks Human NSCs Human embryonic spinal cord Intralesional, injection, 2 × 107 cells ISD Object manipulation, climbing, and over ground manipulation Graft survival over 9 months
Showed axon regeneration with synapse formation
Improved forelimb function
Karova et al. [101] 2019 Rat Thoracic Ballooninduced compression injury 1 Week NPCs Human fetal spinal cord Intralesional, 5 × 105 cells ISD None TNF-α downregulation, p65 NF-κB inhibition
Reduction of glial scar and cavity size
Lien et al. [86] 2019 Rat Cervical Hemisection 2 Weeks Human NSCs Human ESCs Perilesional injections (4 points around lesion cavity), 2.5 × 105 cells, each Growth factor cocktail None No neuron migration
Li et al. [109] 2020 Rat Thoracic Transection 0 Days NSCs Rat fetal brain Perilesional injections (2 points rostral, caudal to lesion), 5 × 105 cells Wnt5a transfection BBB test Wnt5a-induced NSC differentiate into neurons and promote motor functional and histological recovery
Jevans et al. [90] 2021 Rat Thoracic Drop weight contusion injury 3 Days NSCs Rat enteric nervous system Perilesional injection, 1 × 106 cells Chondrotinase ABC Horizontal ladder test Gastrointestinal tract could be a viable option for cell source.
Cotreated with Chondrotinase ABC showed highest regenerative effect with modest functional improvement
Xue et al. [110] 2021 Mouse Thoracic Transection 0 Day NSCs Mouse spinal cord Cord lesion site Collagen nerve regeneration scaffolds BMS score Promotion of neuronal differentiation, synpse formation
Apol8 transfection Improved hindlimb motor function
Epothilone D
Liu et al. [111] 2022 Rat Thoracic Transection 0 Day NSCs Rat fetal brain Cord lesion site 3D bioprinting sodium innateate/gelatin scaffold BBB test Improved hindlimb motor function
OLGs Promoted neural regeneration

SCI, spinal cord injury; NPC, neural progenitor cell; N/A, not available; BBB test, Basso-Beattie-Bresnahan test; NSCs, neural stem cells; PLGA, poly-lactico-glycolic acid; SC, spinal cord; BMS, Basso mouse scale; ISD, immunosuppressant drugs; EGF, epidermal growth factor; bFGF, basic fibroblast growth factor; ESC, embryonal stem cell; PDGF-AA, platelet-derived growth factor; MSC, mesenchymal stem cell; NT-3, neurotrophin-3; 3D, 3-dimensional; OLG, oligodentrocyte; iPC-NP, induced pleuripotent stem cell derived neural precursor cell.

Table 2.
Summary of published clinical trials using neural stem cells in spinal cord injury patients in literature
Study Country Clinical phase Injury location Treatment timing Cell type Cell source Administration route Results
Moviglia et al. [107] 2009 Argentina Phase I Cerivcal/thoracic Chronic* Autologous NSCs Feeding artery infusion Functional recovery was shown in 5/8 patients.
Shin et al. [108] 2015 South Korea Phase I/II Cervical 22–213 days after SCI hNSPCs Human fetal brain Intralesional injection Partial improvements in sensorimotor function
Ghobrial et al. [105] 2017 USA Phase II Cervical/thoracic At least 4 months after SCI NSCs (HuCNS-SC) Human fetal brain Intralesional injection Improvements in overall mean functional outcomes measures
Levi et al. [112] 2018 USA Phase I Cervical/thoracic 4–24 months after SCI NSCs (HuCNS-SC) Human fetal brain Intralesional injection A manual injection technique are safe and feasible
Curtis et al. [113] 2018 USA Phase I Thoracic 1–2 years after SCI NSCs (NSI-566) Human fetal spinal cord Intralesional injection Can be transplanted safely
Levi et al. [106] 2019 USA Phase II Cervical 4–24 months after SCI NSCs (HuCNS-SC) Human fetal brain Intralesional injection Motor functional gains in the treated participants

HuCNS-SC, human fetal-derived central nervous system neural stem cell; NSCs, neural stem cells; NSI-566, NSI-566 cell line human spinalcord-derived neural stem cell; hNSPCs, human neural stem/progenitor cells; SCI, spinal cord injury; USA, United States of America.

* Specific treatment timing after spinal cord injury was not described.

REFERENCES

1. Ahuja CS, Nori S, Tetreault L, et al. Traumatic spinal cord injury-repair and regeneration. Neurosurgery 2017;80:S9-22.
crossref pmid pdf
2. Azarhomayoun A, Aghasi M, Mousavi N, et al. Mortality rate and predicting factors of traumatic thoracolumbar spinal cord injury; a systematic review and meta-analysis. Bull Emerg Trauma 2018;6:181-94.
crossref pmid pmc pdf
3. Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational advances in spinal cord injury. J Neurosurg Spine 2018;30:1-18.
crossref pmid
4. Divanoglou A, Westgren N, Bjelak S, et al. Medical conditions and outcomes at 1 year after acute traumatic spinal cord injury in a Greek and a Swedish region: a prospective, population-based study. Spinal Cord 2010;48:470-6.
crossref pmid pdf
5. Joseph C, Nilsson Wikmar L. Prevalence of secondary medical complications and risk factors for pressure ulcers after traumatic spinal cord injury during acute care in South Africa. Spinal Cord 2016;54:535-9.
crossref pmid pdf
6. Kumar R, Lim J, Mekary RA, et al. Traumatic spinal injury: global epidemiology and worldwide volume. World Neurosurg 2018;113:e345-63.
crossref pmid
7. Choi SH, Sung CH, Heo DR, et al. Incidence of acute spinal cord injury and associated complications of methylprednisolone therapy: a national population-based study in South Korea. Spinal Cord 2020;58:232-7.
crossref pmid pdf
8. Pereira I, Marote A, Salgado A, et al. Filling the gap: neural stem cells as a promising therapy for spinal cord injury. Pharmaceuticals (Basel) 2019;12:65.
crossref pmid pmc
9. Theodore N, Hadley MN, Aarabi B, et al. Prehospital cervical spinal immobilization after trauma. Neurosurgery 2013;72 Suppl 2:22-34.
crossref pmid pdf
10. Li Y, Walker CL, Zhang YP, et al. Surgical decompression in acute spinal cord injury: a review of clinical evidence, animal model studies, and potential future directions of investigation. Front Biol (Beijing) 2014;9:127-36.
crossref pmid pmc pdf
11. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145-7.
crossref pmid
12. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663-76.
crossref pmid
13. Gurusamy N, Alsayari A, Rajasingh S, et al. Adult stem cells for regenerative therapy. Prog Mol Biol Transl Sci 2018;160:1-22.
crossref pmid
14. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143-7.
crossref pmid
15. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells - current trends and future prospective. Biosci Rep 2015;35:e00191.
crossref pmid pmc pdf
16. Gincberg G, Arien-Zakay H, Lazarovici P, et al. Neural stem cells: therapeutic potential for neurodegenerative diseases. Br Med Bull 2012;104:7-19.
crossref pmid
17. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders--time for clinical translation? J Clin Invest 2010;120:29-40.
crossref pmid pmc
18. Gage FH, Temple S. Neural stem cells: generating and regenerating the brain. Neuron 2013;80:588-601.
crossref pmid
19. Gritti A, Parati EA, Cova L, et al. Multipotential stem cells from the adult mouse brain proliferate and self-renew in response to basic fibroblast growth factor. J Neurosci 1996;16:1091-100.
crossref pmid pmc
20. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 1992;255:1707-10.
crossref pmid
21. Obernier K, Alvarez-Buylla A. Neural stem cells: origin, heterogeneity and regulation in the adult mammalian brain. Development 2019;146:dev156059.
crossref pmid pmc pdf
22. Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 2011;70:687-702.
crossref pmid pmc
23. Zhao C, Deng W, Gage FH. Mechanisms and functional implications of adult neurogenesis. Cell 2008;132:645-60.
crossref pmid
24. McIntyre WB, Pieczonka K, Khazaei M, et al. Regenerative replacement of neural cells for treatment of spinal cord injury. Expert Opin Biol Ther 2021;21:1411-27.
crossref pmid
25. Cheng Z, Bosco DB, Sun L, et al. Neural stem cell-conditioned medium suppresses inflammation and promotes spinal cord injury recovery. Cell Transplant 2017;26:469-82.
crossref pmid pmc pdf
26. Gao L, Peng Y, Xu W, et al. Progress in stem cell therapy for spinal cord injury. Stem Cells Int 2020;2020:2853650.
crossref pmid pmc pdf
27. Rong Y, Liu W, Wang J, et al. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. Cell Death Dis 2019;10:340.
crossref pmid pmc pdf
28. Tang Y, Yu P, Cheng L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis 2017;8:e3108.
crossref pmid pmc pdf
29. Suzuki H, Imajo Y, Funaba M, et al. Current concepts of neural stem/progenitor cell therapy for chronic spinal cord injury. Front Cell Neurosci 2021;15:794692.
crossref pmid pmc
30. Vescovi AL, Galli R, Gritti A. The neural stem cells and their transdifferentiation capacity. Biomed Pharmacother 2001;55:201-5.
crossref pmid
31. Farid MF, S Abouelela Y, Rizk H. Stem cell treatment trials of spinal cord injuries in animals. Auton Neurosci 2021;238:102932.
crossref pmid
32. Cummings BJ, Uchida N, Tamaki SJ, et al. Human neural stem cell differentiation following transplantation into spinal cord injured mice: association with recovery of locomotor function. Neurol Res 2006;28:474-81.
crossref pmid
33. Yamazaki K, Kawabori M, Seki T, et al. Clinical trials of stem cell treatment for spinal cord injury. Int J Mol Sci 2020;21:3994.
crossref pmid pmc
34. Galiakberova AA, Dashinimaev EB. Neural stem cells and methods for their generation from induced pluripotent stem cells in vitro. Front Cell Dev Biol 2020;8:815.
crossref pmid pmc
35. Akesson E, Piao JH, Samuelsson EB, et al. Long-term culture and neuronal survival after intraspinal transplantation of human spinal cord-derived neurospheres. Physiol Behav 2007;92:60-6.
crossref pmid
36. Dromard C, Guillon H, Rigau V, et al. Adult human spinal cord harbors neural precursor cells that generate neurons and glial cells in vitro. J Neurosci Res 2008;86:1916-26.
crossref pmid
37. Mothe AJ, Zahir T, Santaguida C, et al. Neural stem/progenitor cells from the adult human spinal cord are multipotent and self-renewing and differentiate after transplantation. PLoS One 2011;6:e27079.
crossref pmid pmc
38. Marei HE, Althani A, Rezk S, et al. Therapeutic potential of human olfactory bulb neural stem cells for spinal cord injury in rats. Spinal Cord 2016;54:785-97.
crossref pmid pdf
39. Amemori T, Romanyuk N, Jendelova P, et al. Human conditionally immortalized neural stem cells improve locomotor function after spinal cord injury in the rat. Stem Cell Res Ther 2013;4:68.
crossref pmid pmc pdf
40. Cheng Z, Zhu W, Cao K, et al. Anti-inflammatory mechanism of neural stem cell transplantation in spinal cord injury. Int J Mol Sci 2016;17:1380.
crossref pmid pmc
41. Karimi-Abdolrezaee S, Eftekharpour E, Wang J, et al. Synergistic effects of transplanted adult neural stem/progenitor cells, chondroitinase, and growth factors promote functional repair and plasticity of the chronically injured spinal cord. J Neurosci 2010;30:1657-76.
crossref pmid pmc
42. Lu P, Wang Y, Graham L, et al. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell 2012;150:1264-73.
crossref pmid pmc
43. Salazar DL, Uchida N, Hamers FP, et al. Human neural stem cells differentiate and promote locomotor recovery in an early chronic spinal cord injury NOD-scid mouse model. PLoS One 2010;5:e12272.
crossref pmid pmc
44. Ma DK, Bonaguidi MA, Ming GL, et al. Adult neural stem cells in the mammalian central nervous system. Cell Res 2009;19:672-82.
crossref pmid pmc pdf
45. Picard-Riera N, Nait-Oumesmar B, Baron-Van Evercooren A. Endogenous adult neural stem cells: limits and potential to repair the injured central nervous system. J Neurosci Res 2004;76:223-31.
crossref pmid
46. Nam H, Lee KH, Nam DH, et al. Adult human neural stem cell therapeutics: current developmental status and prospect. World J Stem Cells 2015;7:126-36.
crossref pmid pmc
47. Maric D, Maric I, Barker JL. Buoyant density gradient fractionation and flow cytometric analysis of embryonic rat cortical neurons and progenitor cells. Methods 1998;16:247-59.
crossref pmid
48. Panchision DM, Chen HL, Pistollato F, et al. Optimized flow cytometric analysis of central nervous system tissue reveals novel functional relationships among cells expressing CD133, CD15, and CD24. Stem Cells 2007;25:1560-70.
crossref pmid pdf
49. Arsenijevic Y, Villemure JG, Brunet JF, et al. Isolation of multipotent neural precursors residing in the cortex of the adult human brain. Exp Neurol 2001;170:48-62.
crossref pmid
50. Johansson CB, Svensson M, Wallstedt L, et al. Neural stem cells in the adult human brain. Exp Cell Res 1999;253:733-6.
crossref pmid
51. Kukekov VG, Laywell ED, Suslov O, et al. Multipotent stem/progenitor cells with similar properties arise from two neurogenic regions of adult human brain. Exp Neurol 1999;156:333-44.
crossref pmid
52. Moe MC, Varghese M, Danilov AI, et al. Multipotent progenitor cells from the adult human brain: neurophysiological differentiation to mature neurons. Brain 2005;128:2189-99.
crossref pmid
53. Palmer TD, Schwartz PH, Taupin P, et al. Cell culture. Progenitor cells from human brain after death. Nature 2001;411:42-3.
crossref pmid pdf
54. Varghese M, Olstorn H, Berg-Johnsen J, et al. Isolation of human multipotent neural progenitors from adult filum terminale. Stem Cells Dev 2009;18:603-13.
crossref pmid
55. Walton NM, Sutter BM, Chen HX, et al. Derivation and large-scale expansion of multipotent astroglial neural progenitors from adult human brain. Development 2006;133:3671-81.
crossref pmid pdf
56. Westerlund U, Moe MC, Varghese M, et al. Stem cells from the adult human brain develop into functional neurons in culture. Exp Cell Res 2003;289:378-83.
crossref pmid
57. Westerlund U, Svensson M, Moe MC, et al. Endoscopically harvested stem cells: a putative method in future autotransplantation. Neurosurgery 2005;57:779-84. discussion 779-84.
crossref pmid
58. Rietze RL, Reynolds BA. Neural stem cell isolation and characterization. Methods Enzymol 2006;419:3-23.
crossref pmid
59. Bez A, Corsini E, Curti D, et al. Neurosphere and neurosphere-forming cells: morphological and ultrastructural characterization. Brain Res 2003;993:18-29.
crossref pmid
60. Reynolds BA, Rietze RL. Neural stem cells and neurospheres---re-evaluating the relationship. Nat Methods 2005;2:333-6.
crossref pmid pdf
61. Suslov ON, Kukekov VG, Ignatova TN, et al. Neural stem cell heterogeneity demonstrated by molecular phenotyping of clonal neurospheres. Proc Natl Acad Sci U S A 2002;99:14506-11.
crossref pmid pmc
62. Joo KM, Kang BG, Yeon JY, et al. Experimental and clinical factors influencing long-term stable in vitro expansion of multipotent neural cells from human adult temporal lobes. Exp Neurol 2013;240:168-77.
crossref pmid
63. Murrell W, Palmero E, Bianco J, et al. Expansion of multipotent stem cells from the adult human brain. PLoS One 2013;8:e71334.
crossref pmid pmc
64. van Gorp S, Leerink M, Kakinohana O, et al. Amelioration of motor/sensory dysfunction and spasticity in a rat model of acute lumbar spinal cord injury by human neural stem cell transplantation. Stem Cell Res Ther 2013;4:57.
crossref pmid pmc pdf
65. Vescovi AL, Reynolds BA, Fraser DD, et al. bFGF regulates the proliferative fate of unipotent (neuronal) and bipotent (neuronal/astroglial) EGF-generated CNS progenitor cells. Neuron 1993;11:951-66.
crossref pmid
66. Fujimoto Y, Abematsu M, Falk A, et al. Treatment of a mouse model of spinal cord injury by transplantation of human induced pluripotent stem cell-derived long-term self-renewing neuroepithelial-like stem cells. Stem Cells 2012;30:1163-73.
crossref pmid pdf
67. Nori S, Okada Y, Yasuda A, et al. Grafted human-induced pluripotent stem-cell-derived neurospheres promote motor functional recovery after spinal cord injury in mice. Proc Natl Acad Sci U S A 2011;108:16825-30.
crossref pmid pmc
68. Nemati SN, Jabbari R, Hajinasrollah M, et al. Transplantation of adult monkey neural stem cells into a contusion spinal cord injury model in rhesus macaque monkeys. Cell J 2014;16:117-30.
pmid pmc
69. Pritchard CD, Slotkin JR, Yu D, et al. Establishing a model spinal cord injury in the African green monkey for the preclinical evaluation of biodegradable polymer scaffolds seeded with human neural stem cells. J Neurosci Methods 2010;188:258-69.
crossref pmid pmc
70. Yamane J, Nakamura M, Iwanami A, et al. Transplantation of galectin-1-expressing human neural stem cells into the injured spinal cord of adult common marmosets. J Neurosci Res 2010;88:1394-405.
crossref pmid
71. Cheng I, Mayle RE, Cox CA, et al. Functional assessment of the acute local and distal transplantation of human neural stem cells after spinal cord injury. Spine J 2012;12:1040-4.
crossref pmid
72. Du BL, Xiong Y, Zeng CG, et al. Transplantation of artificial neural construct partly improved spinal tissue repair and functional recovery in rats with spinal cord transection. Brain Res 2011;1400:87-98.
crossref pmid
73. Hosseini SM, Sani M, Haider KH, et al. Concomitant use of mesenchymal stem cells and neural stem cells for treatment of spinal cord injury: a combo cell therapy approach. Neurosci Lett 2018;668:138-46.
crossref pmid
74. Jin Y, Bouyer J, Shumsky JS, et al. Transplantation of neural progenitor cells in chronic spinal cord injury. Neuroscience 2016;320:69-82.
crossref pmid pmc
75. Kadoya K, Lu P, Nguyen K, et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nat Med 2016;22:479-87.
crossref pmid pmc pdf
76. Kusano K, Enomoto M, Hirai T, et al. Transplanted neural progenitor cells expressing mutant NT3 promote myelination and partial hindlimb recovery in the chronic phase after spinal cord injury. Biochem Biophys Res Commun 2010;393:812-7.
crossref pmid
77. Lu P, Ceto S, Wang Y, et al. Prolonged human neural stem cell maturation supports recovery in injured rodent CNS. J Clin Invest 2017;127:3287-99.
crossref pmid pmc
78. Riemann L, Younsi A, Scherer M, et al. Transplantation of neural precursor cells attenuates chronic immune environment in cervical spinal cord injury. Front Neurol 2018;9:428.
crossref pmid pmc
79. Rosenzweig ES, Brock JH, Lu P, et al. Restorative effects of human neural stem cell grafts on the primate spinal cord. Nat Med 2018;24:484-90.
crossref pmid pmc pdf
80. Salewski RP, Mitchell RA, Shen C, et al. Transplantation of neural stem cells clonally derived from embryonic stem cells promotes recovery after murine spinal cord injury. Stem Cells Dev 2015;24:36-50.
crossref pmid pmc
81. Tashiro S, Nishimura S, Iwai H, et al. Functional recovery from neural stem/progenitor cell transplantation combined with treadmill training in mice with chronic spinal cord injury. Sci Rep 2016;6:30898.
crossref pmid pmc pdf
82. Cheng I, Park DY, Mayle RE, et al. Does timing of transplantation of neural stem cells following spinal cord injury affect outcomes in an animal model? J Spine Surg 2017;3:567-71.
crossref pmid pmc
83. Nguyen HX, Hooshmand MJ, Saiwai H, et al. Systemic neutrophil depletion modulates the migration and fate of transplanted human neural stem cells to rescue functional repair. J Neurosci 2017;37:9269-87.
crossref pmid pmc
84. Pfeifer K, Vroemen M, Caioni M, et al. Autologous adult rodent neural progenitor cell transplantation represents a feasible strategy to promote structural repair in the chronically injured spinal cord. Regen Med 2006;1:255-66.
crossref pmid
85. Xu N, Xu T, Mirasol R, et al. Transplantation of human neural precursor cells reverses syrinx growth in a rat model of post-traumatic syringomyelia. Neurotherapeutics 2021;18:1257-72.
crossref pmid pmc pdf
86. Lien BV, Tuszynski MH, Lu P. Astrocytes migrate from human neural stem cell grafts and functionally integrate into the injured rat spinal cord. Exp Neurol 2019;314:46-57.
crossref pmid
87. Robinson J, Lu P. Optimization of trophic support for neural stem cell grafts in sites of spinal cord injury. Exp Neurol 2017;291:87-97.
crossref pmid
88. Suzuki H, Ahuja CS, Salewski RP, et al. Neural stem cell mediated recovery is enhanced by Chondroitinase ABC pretreatment in chronic cervical spinal cord injury. PLoS One 2017;12:e0182339.
crossref pmid pmc
89. Nori S, Khazaei M, Ahuja CS, et al. Human oligodendrogenic neural progenitor cells delivered with chondroitinase ABC facilitate functional repair of chronic spinal cord injury. Stem Cell Reports 2018;11:1433-48.
crossref pmid pmc
90. Jevans B, James ND, Burnside E, et al. Combined treatment with enteric neural stem cells and chondroitinase ABC reduces spinal cord lesion pathology. Stem Cell Res Ther 2021;12:10.
crossref pmid pmc pdf
91. Günther MI, Weidner N, Müller R, et al. Cell-seeded alginate hydrogel scaffolds promote directed linear axonal regeneration in the injured rat spinal cord. Acta Biomater 2015;27:140-50.
crossref pmid
92. Huang L, Wang Y, Zhu M, et al. Anisotropic alginate hydrogels promote axonal growth across chronic spinal cord transections after scar removal. ACS Biomater Sci Eng 2020;6:2274-86.
crossref pmid
93. Yin W, Xue W, Zhu H, et al. Scar tissue removal-activated endogenous neural stem cells aid Taxol-modified collagen scaffolds in repairing chronic long-distance transected spinal cord injury. Biomater Sci 2021;9:4778-92.
crossref pmid
94. Lopez-Dolado E, Gonzalez-Mayorga A, Gutierrez MC, et al. Immunomodulatory and angiogenic responses induced by graphene oxide scaffolds in chronic spinal hemisected rats. Biomaterials 2016;99:72-81.
crossref pmid
95. Gelain F, Panseri S, Antonini S, et al. Transplantation of nanostructured composite scaffolds results in the regeneration of chronically injured spinal cords. ACS Nano 2011;5:227-36.
crossref pmid
96. Olson HE, Rooney GE, Gross L, et al. Neural stem cell- and Schwann cell-loaded biodegradable polymer scaffolds support axonal regeneration in the transected spinal cord. Tissue Eng Part A 2009;15:1797-805.
crossref pmid pmc
97. Amemori T, Ruzicka J, Romanyuk N, et al. Comparison of intraspinal and intrathecal implantation of induced pluripotent stem cell-derived neural precursors for the treatment of spinal cord injury in rats. Stem Cell Res Ther 2015;6:257.
crossref pmid pmc pdf
98. Cheng I, Githens M, Smith RL, et al. Local versus distal transplantation of human neural stem cells following chronic spinal cord injury. Spine J 2016;16:764-9.
crossref pmid
99. Yousefifard M, Rahimi-Movaghar V, Nasirinezhad F, et al. Neural stem/progenitor cell transplantation for spinal cord injury treatment; a systematic review and meta-analysis. Neuroscience 2016;322:377-97.
crossref pmid
100. Xiong LL, Zou Y, Shi Y, et al. Tree shrew neural stem cell transplantation promotes functional recovery of tree shrews with a hemisectioned spinal cord injury by upregulating nerve growth factor expression. Int J Mol Med 2018;41:3267-77.
crossref pmid pmc
101. Karova K, Wainwright JV, Machova-Urdzikova L, et al. Transplantation of neural precursors generated from spinal progenitor cells reduces inflammation in spinal cord injury via NF-kappaB pathway inhibition. J Neuroinflammation 2019;16:12.
crossref pmid pmc
102. Kumamaru H, Saiwai H, Kubota K, et al. Therapeutic activities of engrafted neural stem/precursor cells are not dormant in the chronically injured spinal cord. Stem Cells 2013;31:1535-47.
crossref pmid pdf
103. Bozkurt G, Mothe AJ, Zahir T, et al. Chitosan channels containing spinal cord-derived stem/progenitor cells for repair of subacute spinal cord injury in the rat. Neurosurgery 2010;67:1733-44.
crossref pmid
104. Nomura H, Zahir T, Kim H, et al. Extramedullary chitosan channels promote survival of transplanted neural stem and progenitor cells and create a tissue bridge after complete spinal cord transection. Tissue Eng Part A 2008;14:649-65.
crossref pmid
105. Ghobrial GM, Anderson KD, Dididze M, et al. Human neural stem cell transplantation in chronic cervical spinal cord injury: functional outcomes at 12 months in a phase II clinical trial. Neurosurgery 2017;64:87-91.
crossref pmid
106. Levi AD, Anderson KD, Okonkwo DO, et al. Clinical outcomes from a multi-center study of human neural stem cell transplantation in chronic cervical spinal cord injury. J Neurotrauma 2019;36:891-902.
crossref pmid
107. Moviglia GA, Varela G, Brizuela JA, et al. Case report on the clinical results of a combined cellular therapy for chronic spinal cord injured patients. Spinal Cord 2009;47:499-503.
crossref pmid pdf
108. Shin JC, Kim KN, Yoo J, et al. Clinical trial of human fetal brain-derived neural stem/progenitor cell transplantation in patients with traumatic cervical spinal cord injury. Neural Plast 2015;2015:630932.
crossref pmid pmc pdf
109. Li X, Peng Z, Long L, et al. Transplantation of Wnt5a-modified NSCs promotes tissue repair and locomotor functional recovery after spinal cord injury. Exp Mol Med 2020;52:2020-33.
crossref pmid pmc pdf
110. Xue W, Zhang H, Fan Y, et al. Upregulation of Apol8 by Epothilone D facilitates the neuronal relay of transplanted NSCs in spinal cord injury. Stem Cell Res Ther 2021;12:300.
crossref pmid pmc pdf
111. Liu S, Yang H, Chen D, et al. Three-dimensional bioprinting sodium alginate/gelatin scaffold combined with neural stem cells and oligodendrocytes markedly promoting nerve regeneration after spinal cord injury. Regen Biomater 2022;9:rbac038.
crossref pmid pmc pdf
112. Levi AD, Okonkwo DO, Park P, et al. Emerging safety of intramedullary transplantation of human neural stem cells in chronic cervical and thoracic spinal cord injury. Neurosurgery 2018;82:562-75.
crossref pmid pdf
113. Curtis E, Martin JR, Gabel B, et al. A first-in-human, phase I study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell 2018;22:941-50. e6.
crossref pmid


Editorial Office
CHA University, CHA School of Medicine Bundang Medical Center
59 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13496, Korea
Tel: +82-31-780-1924  Fax: +82-31-780-5269  E-mail: support@e-neurospine.org
The Korean Spinal Neurosurgery Society
#407, Dong-A Villate 2nd Town, 350 Seocho-daero, Seocho-gu, Seoul 06631, Korea
Tel: +82-2-585-5455  Fax: +82-2-2-523-6812  E-mail: ksns1987@gmail.com
Business License No.: 209-82-62443

Copyright © The Korean Spinal Neurosurgery Society.

Developed in M2PI

Close layer
prev next