You are on page 1of 17

Biomaterials 279 (2021) 121211

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Stem cell-derived neuronal relay strategies and functional electrical


stimulation for treatment of spinal cord injury
Bi-Qin Lai a, d, Xiang Zeng a, Wei-Tao Han b, Ming-Tian Che a, Ying Ding b, Ge Li a,
Yuan-Shan Zeng a, b, c, d, e, *
a
Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
b
Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
c
Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
d
Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
e
Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan, School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China

A R T I C L E I N F O A B S T R A C T

Keywords: The inability of adult mammals to recover function lost after severe spinal cord injury (SCI) has been known for
Neuronal relay millennia and is mainly attributed to a failure of brain-derived nerve fiber regeneration across the lesion. Po­
Functional electrical stimulation tential approaches to re-establishing locomotor function rely on neuronal relays to reconnect the segregated
Stem cells
neural networks of the spinal cord. Intense research over the past 30 years has focused on endogenous and
Spinal cord injury
Neural network
exogenous neuronal relays, but progress has been slow and the results often controversial. Treatments with stem
cell-derived neuronal relays alone or together with functional electrical stimulation offer the possibility of
improved repair of neuronal networks. In this review, we focus on approaches to recovery of motor function in
paralyzed patients after severe SCI based on novel therapies such as implantation of stem cell-derived neuronal
relays and functional electrical stimulation. Recent research progress offers hope that SCI patients will one day be
able to recover motor function and sensory perception.

1. Introduction repair using a neuronal relay has gradually been established and vali­
dated [9–12]. Several studies have proposed use of neuronal relay-based
Severe spinal cord injury (SCI), such as complete spinal cord tran­ repair strategies to provide endogenous or exogenous neuronal relays
section manifested by paraplegia or quadriplegia, causes tremendous (for example, neuronal network or spinal cord-like tissue), while
physical pain and mental anguish for patients while placing a heavy improving the microenvironment within the damaged spinal cord tissue
financial burden on the patient’s family and society as a whole [1,2]. In [8,13–15]. These strategies use novel tissue engineering technologies
spite of hundreds of years of research into SCI repair, no breakthrough involving stem cells, cytokines, and biomaterials. They are able to
has yet been made, and clinical demand for SCI treatment continues to receive ascending and descending neural information, and to transmit
grow [3–5]. In order to improve quality of life for this population and neural information to propriospinal nerve endings at both ends of the
reduce the social burden, development of effective treatment strategies injured area. SCI repair using neuronal relays has transformed the
for SCI is needed. This will require a better understanding of processes traditional idea that “ascending and descending nerve fibers in adult
underlying the repair of severe SCI at both the structural and functional mammalian spinal cord must regenerate across the SCI area with entire
levels [6]. complete spinal cord transection to restore spinal cord motion and
Severe SCI in adult mammals generally makes it difficult for sensory function” [16–19]. In this review, we focus on recent progress in
descending nerve fibers in the corticospinal tract (CST) to achieve long- research on SCI repair using stem cell-derived neuronal relay strategies
distance across the injured area, resulting in difficulty in restoring distal and functional electrical stimulation, both of which use novel interdis­
motor function [7,8]. In the more than 30 years since the first trans­ ciplinary biomedical technologies to achieve repair of motor and sen­
plantation of embryonic neural tissue into an SCI site, the concept of SCI sory function after complete SCI.

* Corresponding author. Department of Histology and Embryology Zhongshan School of Medicine Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou,
510080, China.
E-mail address: zengysh@mail.sysu.edu.cn (Y.-S. Zeng).

https://doi.org/10.1016/j.biomaterials.2021.121211
Received 29 December 2020; Received in revised form 9 October 2021; Accepted 20 October 2021
Available online 22 October 2021
0142-9612/© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

2. Challenges of SCI repair 3. Neuronal relay function

Tissue ischemia, necrosis, edema, and oxidative stress during the Although brain-derived descending nerve fibers such as those in the
acute phase of SCI can result in neuronal and glial cell necrosis and CST cannot easily regenerate across the SCI area to reconnect with target
apoptosis. Over time, the injured area continues to expand, forming a neurons, it may be feasible to provide an interneuronal network con­
microenvironment that is unfavorable to nerve regeneration owing to taining “hand-in-hand connections” for relaying neural information
processes such as inflammatory factor infiltration, glial scarring, fibrous through the defective area [12,16,39]. This interneuronal network could
scarring, and cavity formation [20–22]. Post-SCI structural and func­ perform an information-relaying role in the SCI area in the following
tional reconstruction is a challenge for both basic and applied research. ways (Fig. 1). In one direction, it would establish synaptic connections
Problems to be urgently addressed include: (1) how to improve the with nerve fibers transmitting brain-derived descending motor neuronal
post-injury microenvironment and reconstruct the damaged neural signals and would deliver these signals to motor neurons in the caudal
network after SCI [21,23], (2) how to replace dead neurons and oligo­ area to the injury site of spinal cord. In the other direction, it would
dendrocytes [24], and (3) how to integrate new functional cells into the establish synaptic connections with nerve fibers transmitting informa­
host neural network to repair the structure and restore the function of tion carried by sensory nerve fibers and upload this information to the
the injured spinal cord [25]. Solving these problems depends on brainstem and the cerebrum. This would enable the higher central
in-depth knowledge of the mechanisms of SCI repair. nervous system to regulate the accuracy of limb movement according to
SCI can be subcategorized into incomplete and complete SCI. A series the afferent sensory signals it receives [35,40]. However, in order for
of experiments have shown that early adoption of specific strategies these neuronal relays to become functional, it is necessary for synapses
(such as reducing tissue edema, inhibiting inflammation, providing to form between the host axons extending from the rostral area and relay
neurotrophic factors, and carrying out rehabilitation therapy after neurons at the injury site (Fig. 1). To establish functionality, neuro­
incomplete SCI) can facilitate reconnection of residual damaged neurons transmitters must also be appropriately released from the relay neurons,
via collateral sprouting of adjacent undamaged axons [21,26–29]. Use and axons must extend from them to form synapses with neurons in
of small molecule inhibitors to reduce the excitability of inhibitory in­ areas that are caudal to the injury site [33,39,41]. Finally, axons that
terneurons in the injured spinal cord enhances responsiveness to traverse the lesion need to undergo remyelination [42,43]. Recent
descending excitatory signals and promotes functional recovery after studies have demonstrated that neuronal relays can be formed from
SCI in mice with staggered bilateral hemisections in which the lum­ various cells, and can be applied to a range of SCI models with the aim of
barspinal cord is deprived of all direct brain-derived innervation while restoring motor, sensory, and autonomic function [8,13,19]. However,
retaining dormant relay circuits [30]. These findings suggest that by it is important that efforts to refine these techniques continue in order to
establishing new synaptic connections with relay neurons, the improve the specificity of neuronal connections while enhancing func­
brain-derived descending neural information is retransmitted to target tional outcomes and reproducibility.
neurons that regulate limb movement, thereby achieving functional An increasing number of researchers are attempting to use this relay
repair [16,31]. However, in patients with paraplegia or quadriplegia strategy to provide an interneuronal network at the SCI site to re-
caused by complete SCI, such reconnection via axonal collateral establish neural transmission through the spinal cord (Table 1). Based
sprouting is precluded; all axon bundles must traverse the lesion area. on research studies and the neuronal relay strategy most commonly used
Even using a combination of treatments, it is still difficult to regenerate in recent years (Fig. 2), the majority of researchers have attributed the
brain-derived descending nerve fibers, especially in the CST that is behavioral recovery of animals after complete SCI to synaptic connec­
responsible for governing autonomic motor function. This is mainly due tions between regenerating propriospinal nerve fibers and neurons
to microenvironmental factors in the area of SCI inhibiting nerve transplanted into the injury site [21,51,59]. These donor neurons act as
regeneration and the inability of neuronal axons to dynamically regen­ interneurons to relay neural information through the spinal cord neural
erate [4,32]. In the past decade, researchers have made many attempts network, and are either excitatory or inhibitory [17,21,59]. Although
to improve the microenvironment of the SCI site (including strength­ there are no reports to date on whether a particular type of neuron acts
ening the axonal regeneration [27,33], transplanting and replacing cells as a relay interneuron, Lai et al. [35] did confirm in their previous
[15], and administering gene therapy [34]) and have found that mouse studies that excitatory interneurons play a major role in recovery of
CST nerve fibers can at least partially regenerate. However, in large motor function. The conceptual focus of this study was whether relay
mammals, the truncated bundles at the CST site are unable to suffi­ neurons are able to transmit information via the host axon in a form that
ciently regenerate across the SCI area to reconnect the target neurons at is actually related to the desired motor signals. The data showed that
the end of the injury site [8,19]. more than half of the donor neurons were capable of synthesizing
In addition to the absence of CST regeneration, the inability to excitatory neurotransmitters and could integrate with host axons to
restore motor function after SCI may also be associated with a lack of transmit descending excitatory signals from the brain. Furthermore, the
regeneration of other descending nerve fibers that project to the spinal axons of donor neurons extended to the adjacent caudal segment of the
cord. These descending nerve fibers include the rubrospinal tract (RST), host tissue and successfully formed synaptic connections with the host
reticulospinal tract, 5-hydroxytryptamine (5-HT)-positive nerve fibers, propriospinal neurons via excitatory presynaptic buttons. These mech­
tyrosine hydroxylase (TH)-positive nerve fibers, dopaminergic nerve anisms enabled the experimental animals to regain weight-bearing
fibers, and spinal cord fasciculus proprius nerve fibers [2,35,36]. Some locomotion and also to coordinate their stepping movements.
studies suggest that the brain-derived 5-HT- and TH-positive nerve fibers The role of inhibitory relay neurons is also not negligible, given that
are more likely than CST nerve fibers to regenerate into sites of SCI, canines with transplanted inhibitory relay neurons have shown signs of
which is an important factor for improving motor function after SCI coordinated motor recovery [35]. Our view is that there are changes in
[35–37]. Moreover, regeneration of ascending nerve fibers is essential synaptic plasticity as new relay neurons integrate into the propriospinal
for restoration of locomotor coordination [38]. Therefore, it is necessary neural network; that is, the spinal cord is able to retain functional neural
to fully activate regeneration of brain-derived nerve fibers and sensory pathways in response to physical therapy or rehabilitation training
nerve fibers when repairing spinal nerve circuits. Adequate consider­ [60–63]. In the process, as the survival time of stem cell-derived donor
ation should be given to effective integration of neuronal relay, neurons at the injury/graft site of spinal cord increases, those neurons
ascending and descending nerve fibers. and their axons that do not form functional connections are removed,
leaving only those donor relay neurons that can establish functional
connections with the host neurons. Some researchers have used a
counterevidence strategy, opting to ablate the neural network at the SCI

2
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Fig. 1. Schematic diagram of neuronal relay forma­


tion models for the transmission of descending and
ascending neural signals in SCI repair. A) Relay neu­
rons (green) might receive and integrate neural sig­
nals from multiple descending nerve fibers and output
these to propriospinal neurons in the injury site of the
completely transected spinal cord. B) Relay neurons
(green) may also receive and integrate neural signals
from multiple ascending nerve fibers and output these
to supraspinal neurons and propriospinal neurons.
DRG, dorsal root ganglion.

Table 1
Summary of neuronal relay strategies in animal models of complete spinal cord transection.
Strategies Target cells in the injury/ Important principles Challenges Refs
graft site

Guiding endogenous neurogenesis Endogenous NSCs Inducing neuronal differentiation of Insufficient endogenous NSCs or NSCs [12,34,44,
endogenous NSCs depleted 45]
In vivo direct reprogramming Cells in injured spinal cord Converting local cells directly into neurons Low reprogramming efficiency [46–48]
tissue or glial scar tissue
Cell transplantation Stem cells or NPCs Inducing neuronal differentiation of Long-distance cell migration and [15,41,
exogenous stem cells or NPCs colony formation 49–52]
Tissue-engineered neural network Gene-modified cells in a Integrating donor functional neurons with the Long-term survival and functional [19,25,35,
tissue or spinal cord-like tissue engineered tissue implant host propriospinal neural network integration with the host spinal cord 37,53–55]
transplantation tissue
Fetal spinal cord tissue transplantation Neural cells in a fetal spinal Providing fetal neurons, oligodendrocytes, Sourcing and ethical limitations in [9,10,
cord tissue implant astrocytes, NSCs, NPCs and the extracellular translational medicine 56–58]
matrix

Abbreviations: NSCs, neural stem cells; NPCs, neural precursor cells.

site and have found that motor function in the animals is again impaired neurotrophic factors, the use of specially modified materials that allow
[51,64,65], which functionally confirms the existence of the neuronal the slow release of neurotrophic factors, or the use of drugs that inhibit
relay. Other researchers have confirmed the existence of the neuronal the inflammatory response or promote angiogenesis at the site of injury.
relay at the structural and functional levels using trans-synaptic nerve Moreover, repair attempts are made not only to support stem
tracing, immunoelectron microscopy, and neurophysiological tech­ cell-derived interneuron survival, but also to promote the formation of
niques [35,37]. synaptic connections between the propriospinal regenerating nerve fi­
It is worth noting that, in adopting a neuronal relay strategy to repair bers and the interneurons at the SCI site, as well as to increase remye­
SCI, simple supplementation of stem cell-derived interneurons has lination [26,43,59]. Only in this way can the newly replenished stem
yielded unsatisfactory results, so the combination of multiple factors has cell-derived interneurons act effectively as a relay. Besides, before the
to be considered. Ensuring the success of the interneuronal relay neuronal relay can be effective, the atrophy, functional silencing, and
network at the SCI site requires improving the adverse microenviron­ apoptosis of the denervated spinal cord neurons or muscle has to be
ment of the injured area, as this microenvironment is not conducive to prevented [30,67]. It is necessary to consider whether rehabilitation
the survival and integration of the network [20,66]. The latest ap­ training and physiotherapy can be used in combination with a neuronal
proaches for improving the adverse conditions of the specific microen­ relay strategy to promote the formation of a greater number of correct
vironment at the site of injury include the simple addition of synaptic connections between the regenerating nerve fibers and the stem

3
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Fig. 2. Therapeutic strategies for neuronal relays. A) Transplantation of a bioactive factor-modified scaffold into the injury site in the spinal cord attracts migrating
endogenous stem cells and promotes their differentiation into relay neurons. B) Tissue-engineered neural network tissue is transplanted into the injury site in the
spinal cord, and plays a role in the neuronal relay via the relay neurons. C) Stem cells are directly injected into the injury site of spinal cord and induced to
differentiate into neurons. D) Transcription factors are directly injected into the injured spinal cord tissue and induce the reprogramming of glial cells into neurons.

cell-derived interneurons [68]. spinal cords in which function was restored by replacing spinal cord
segments which then successfully connected with host spinal cord. In
this neonatal rat injury model, transplantation of embryonic spinal cord
3.1. Endogenous neuronal relays tissue promoted robust growth and regrowth of axons across the graft,
resulting in nearly normal neural connections. Animals that had been
As early as the 1980s, it was demonstrated that embryonic spinal fitted with replacement segments were able to walk, run, and climb with
cord tissue transplanted into the SCI site can survive and promote levels of hind-forelimb coordination that were almost normal. However,
regeneration of brain-derived descending nerve fibers [9,10,56]. These given the strict ethical limitations on research in humans and the diffi­
regenerating nerve fibers (i.e., CST nerve fibers) are capable of estab­ culty of obtaining embryonic nerve tissue, this strategy cannot meet the
lishing synaptic connections with transplanted embryonic neurons, clinical demands of SCI treatment. Owing to the continued rapid
leading to improvement of motor function. The concept of an early advancement in biomedical technology, scientists are beginning to
neuronal relay was also introduced at that time [12,16–18,39]. Impor­ develop neuronal relay strategies that are more suitable for clinical
tantly, Reier et al. [9] reported their findings regarding transplantation applications.
of fetal rat spinal cord tissue into 2–4 mm-long intraspinal cavities in Based on the concept of creating an endogenous neuronal relay at the
both adult and neonatal rats. The authors identified surviving donor SCI site (i.e., via activation and induction of neuronal differentiation
tissues and studied their differentiation and extent of fusion with host from endogenous stem cells to form a neural network), Li and colleagues
spinal cords. The data showed that over 80% of the grafts survived in the investigated a biodegradable neurotrophin-3 (NT-3)-loaded chitosan
lesion sites in the spinal cords of both neonatal and adult rats for periods material that could provide 14 weeks of controlled NT-3 release [44].
of 1–16 months. Furthermore, the authors provided evidence that the They found that transplantation of this material improved the micro­
donor tissue had undergone maturation, including demonstrating environment at the SCI site, resulting in regeneration of long-distance
certain topographical features that are characteristic of normal spinal nerve fibers and recruitment of endogenous neural stem cells (NSCs).
cord. Many of the transplants extended over the entire length of the These NSCs were found to proliferate and migrate into the injury/graft
lesion and were often closely apposed to the injured surfaces of the host site in the spinal cord, where they were able to further differentiate into
spinal cords without an intervening dense glial scar. In addition, the interneurons. Such stem cell-derived interneurons were able to establish
authors identified neurons and axons within the graft using retrograde connections with propriospinal neurons and to transmit ascending and
and anterograde labeling, respectively. Taken together, these results descending neural signals to proper targets, ultimately improving motor
demonstrate a clear potential for transplanting embryonic spinal cord function in experimental animals [44,45,69,70]. Dai and colleagues
tissue to replace damaged intraspinal neuronal populations in mammals. developed a functional collagen-based scaffold that could bind to
In another study, Iwashita et al. [57] reported reconstruction of severed

4
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

different growth factors and was able to significantly promote endoge­ vivo studies showed that Zfp521 treatment also affects endogenous as­
nous neurogenesis at the SCI site and improve motor function in rats and trocytes and thus promotes further functional recovery following
canines [71–74]. In one study, the same researchers investigated the experimental SCI in rats. The authors further reported that Zfp521 is
effect of the microtubule-stabilizing agent paclitaxel (PTX) on the able to reprogram cultured astrocytes more efficiently than SOX2, and
endogenous differentiation of NSCs. They loaded PTX-encapsulated li­ that resident astrocytes within the injured spinal cord of adult rats can
posomes into a collagen microchannel scaffold, which released PTX in a be reprogrammed by Zfp521 into neurons via a neuronal progenitor
prolonged and sustained manner when it was implanted at the complete stage. Treatment with Zfp521 led to a significant enhancement in hind
transection spinal cord injury site in a rat model. This scaffold provided a limb locomotor activity, step length, toe spread, foot length, and paw
microenvironment that was suitable for neuronal differentiation of NSCs area. These reprogramming strategies have been shown to induce a high
and the extension of neuronal axons. Endogenous neurogenesis at the rate of neuronal differentiation in vitro. However, due to the influence of
site of spinal cord injury eventually led to improvements in both the complex, damaged microenvironment of the injured spinal cord,
motor-evoked potentials and recovery of hindlimb locomotion. In reprogramming efficiency is lower in vivo than in vitro [79–81].
another study, the same researchers investigated the effect of the Therefore, to date, such reprogramming strategies have seldom been
microtubule-stabilizing agent PTX on the endogenous differentiation of used for complete SCI repair (Table 2) [47,82,83].
NSCs. They loaded PTX-encapsulated liposomes into a collagen micro­ Analysis of papers reporting on endogenous neuronal relay strategies
channel scaffold, which released PTX in a prolonged and sustained published in recent years shows that the ependymal cells of the central
manner when it was implanted at the complete transection spinal cord canal of the spinal cord have NSC-like potential in different mammals
injury site in a rat model. This scaffold provided a microenvironment ranging from rodents to primates [69,76,90,91]. These cells are not
that was suitable for neuronal differentiation of NSCs and the extension active in normal spinal cord tissue, but become activated in response to
of neuronal axons. Endogenous neurogenesis at the site of spinal cord injury and migrate from the central canal into the injury site. In the
injury eventually led to improvements in both motor-evoked potentials microenvironment of the injury site, these activated ependymal cells
and recovery of hindlimb locomotion. Han et al. [75] transplanted a tend to differentiate into astrocytes rather than neurons. These astro­
linear-ordered collagen scaffold bound to brain-derived neurotrophic cytes accumulate at the edge of the lesion and form a dense glial scar,
factor (BDNF) (using a tagged collagen-binding domain) into a canine hindering nerve fiber regeneration [32,90,92–94]. Several bioactive
model of complete spinal cord transection. This linear-ordered collagen factors have been shown to be capable of ameliorating this adverse
scaffold-BDNF complex was shown to effectively promote generation, microenvironment, effectively promoting activation of ependymal cells
maturation, and synaptic formation, as well as axonal remyelination, of with NSC-like potential that have migrated into the site of spinal cord
endogenous stem cell-derived interneurons at the SCI/graft site, thereby injury (Fig. 2) [44,73,95]. Furthermore, these ependymal cells induce
restoring motor function. In addition to the studies described above, the NSC-like cells to differentiate into interneurons that eventually serve
several have also shown that manipulation of the SCI microenvironment a relay role during repair of the spinal cord neuronal network [66,76].
in order to guide endogenous neurogenesis can promote recovery of Some studies have demonstrated that endogenous NSC-derived neurons
motor function in animals with spinal cord deletions [60,76]. function as relay neurons by receiving supraspinal input and extend
Collectively, these studies demonstrate that a fundamental principle their axons across the injury/graft site into the caudal spinal cord for
underlying formation of neuronal relays at sites of injury, along with connectivity [28,96,97]. Even so, before using bioactive factors, at­
recovery of motor function, is initiation of endogenous neurogenesis tempts should be made to fully consider the time course of endogenous
from NSCs. Furthermore, these studies suggest that formation of stem cell activation after SCI. Previous studies have revealed that
neuronal relays, as well as recovery of function, may be more successful NSC-like cells in rat spinal cord reach their peak of activation on the fifth
if material could be implanted at the injury site that can create a pro- day after injury [12,73]. Therefore, the use of bioactive factors depends
regenerative microenvironment to facilitate formation of NSC progeny on the particular time course of endogenous stem cell activation after
and ultimately a greater number of relay neurons. These results SCI in different animals. In addition, the influence of the chosen carrier
emphasize that the primary mechanism underlying regenerative repair and mechanism of bioactive factor release on the effectiveness of this
in SCI is differentiation of endogenous NSCs into interneurons that then strategy should be considered. If bioactive factors can be loaded into a
function as neuronal relays. Thus, when designing endogenous neuronal hydrogel, they can be injected directly into the injured area of the spinal
relay treatments for animals with complete SCI, it is essential that cord [28,97,98]. Some bioactive factors can be loaded into a collagen
neuronal relay function be established that results in formation of scaffold and slowly released during collagen degradation to exert a
appropriate axonal reconnections with the original downstream targets long-lasting effect (Fig. 2) [66,70,86]. More importantly, in addition to
[12,44,69]. activating endogenous stem cells and allowing them to migrate into
Another novel strategy for promoting the growth of endogenous in­ injured spinal cord tissue to differentiate into interneurons, there is
terneurons at the site of SCI is to reprogram glial cells (e.g., astrocytes) to another key issue to consider when attempting to improve motor func­
differentiate into interneurons in vivo (Fig. 2) [46–48,77]. An early tion. This is whether bioactive factors and their carriers can effectively
study established that the adult mammalian spinal cord lacks intrinsic support survival and functional maturation of endogenous stem
neurogenic capacity [78]. On the basis of this finding, Wang et al. [78] cell-derived interneurons, as well as formation of new synaptic con­
attempted to generate neurons after SCI by reprogramming glial cells in nections between these interneurons and regenerating nerve fibers. A
vivo. The authors showed that the p53-dependent signaling pathway promising research direction is to find a combination of bioactive factors
constitutes a critical checkpoint for SOX2-mediated reprogramming of that can inhibit inflammation while promoting nerve fiber regeneration
resident glial cells in the adult mouse spinal cord. They also found that and differentiation of endogenous NSCs into relay interneurons. Dif­
the p53 pathway promoted cell-cycle exit in SOX2-induced adult neu­ ferentiation of endogenous NSCs into oligodendrocytes to repair
roblasts (iANBs). As such, silencing of either p53 or p21 led to a sig­ demyelinated axons also plays an important role in motor and sensory
nificant increase in the overall production of iANBs. Furthermore, BDNF repair. Moreover, a potentially valuable research direction with many
and Noggin created a neurotrophic milieu that promoted maturation of applications would be to develop a scaffold that loads bioactive factors
iANBs into a diverse range of neuronal cell types, although gluta­ to allow for long-term stable release and provides directional guidance
matergic neurons predominated. The study identified critical molecular for nerve fiber regeneration and remyelination (Fig. 2) [99,100]. It is
and cellular checkpoints that can be manipulated to enhance neuro­ important to consider that while therapeutic strategies based on
genesis after SCI. In another study, Zarei et al. [48] demonstrated that endogenous stem cell activation may have better effects for acute SCI, in
ectopic expression of zinc-finger transcription factor 521 (Zfp521) re­ cases of chronic SCI, aging patients with SCI, complete SCI with stem cell
programs fibroblasts and astrocytes into NSCs in vitro. Additional in depletion or insufficient mobilization, it may be necessary to adopt a

5
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Table 2
Summary of endogenous neuronal relay strategies and their effectiveness for complete SCI repair.
Publication Species Treatment Autonomous Electrophysi- Key outcomes in the injury/graft site of Synapselike
locomotor ological spinal cord structure in the
recovery improvement injury/graft site

2006. Ohori Rat Injection of growth factor and NR NR Replacing lost neurons and Yes
et al. [34] transcription factors oligodendrocytes
2013. Li et al. Rat Implantation of EGFR antibody- Yes NR Inducing differentiation of endogenous Yes
[71] functionalized collagen scaffolds NPCs into neurons
2014. Su et al. Mouse Injection of lentivirus NR NR Converting local astrocytes into synapse- Yes
[46] forming interneurons
2014. Guo et al. Mouse Injection of lentiviral vector Yes NR Activating and inducing endogenous NSC NR
[84] containing Sox11 differentiation into neurons, and inducing
expression of BDNF
2015. Han et al. Canine Implantation of linear-ordered Yes Yes Promoting nerve regeneration and NR
[75] collagen binding BDNF improving myelination
2015. Yang et al. Rat Implantation of NT-3-coupled Yes Yes Promoting endogenous neurogenesis Yes
[44] chitosan
2016. Li et al. Rat Implantation of collagen scaffold Yes NR Reducing cavity volume, facilitating nerve NR
[72] and administration of cAMP regeneration, and promoting neurogenesis
2017. Yang et al. Mouse Injection of substance P Yes NR Activating endogenous NSCs and enhancing NR
[85] neurogenesis
2017. Li et al. Canine Implantation of cetuximab- Yes Yes Activating endogenous neurogenesis and Yes
[73] modified collagen scaffold reducing chondroitin sulfate proteoglycan
deposition
2017. Fan et al. Rat Implantation of EGFR antibody Yes Yes Blocking myelin-associated inhibitory Yes
[86] CBD-Fab-modified collagen molecules, guiding nerve regeneration, and
scaffold enhancing neurogenesis
2016/2018. Li Canine Implantation of NT-3/fibroin- Yes Yes Promoting nerve regeneration, attenuating Yes
et al. [60,87] coated gelatin sponge scaffold inflammation, and attracting migrating
cells
2018. Rao et al. Monkey Implantation of NT-3-loaded Yes Yes Promoting nerve regeneration NR
[45] chitosan biodegradable material
2018. Zhang Rat Injection of erythropoietin Yes NR Promoting neurogenesis and NR
et al. [24] oligodendrogenesis
2018. Li et al. Rat Implantation of a collagen Yes Yes Inducing differentiation of endogenous NR
[74] microchannel scaffold carrying NSCs into neurons
paclitaxel-liposomes
2018. Zhou et al. Mouse Implantation of a conducting Yes NR Activating endogenous NSCs and eliciting NR
[88] polymer hydrogel neurogenesis
2019. Zarei- Rat Injection of zinc-finger Yes Yes Converting astrocytes into neuron-like cells NR
Kheirabadi transcription factor
et al. [48]
2019. Shang Rat Implantation of bFGF-Sodium Yes NR Reducing microglial activation, increasing NR
et al. [66] hyaluronate collagen scaffold angiogenesis, eliciting endogenous
neurogenesis, and promoting nerve
regeneration
2019. Pan et al. Rat Implantation of PLGA/GO Yes Yes Increasing neurons survival NR
[23] electrospun nanofibers
containing immobilized IGF-1
and BDNF
2019. Sperling Rat Injection of galantamine Yes NR Increasing neurogenesis and tissue cell NR
et al. [89] survival
2020. Ma et al. Mouse Injection of photo-crosslinked Yes Yes Reducing inflammation and inducing NR
[29] hydrogel and CSF1R inhibitor differentiation of endogenous NPCs into
neurons

Abbreviations: NR, not reported; EGFR, epidermal growth factor receptor; NPCs, neural precursor cells; Sox11, sex-determining region Y-box 11; NSCs, neural stem
cells; BDNF, brain-derived neurotrophic factor; NT-3, neurotrophin-3; cAMP, cyclic adenosine monophosphate; CBD, collagen-binding domain; Fab, Fab fragment of
antibody; bFGF, basic fibroblast growth factor; GO, graphene oxide; PLGA, poly(lactic-co-glycolic acid); IGF-1, insulin-like growth factors-1; CSF1R, colony stimulating
factor-1 receptor.

therapeutic strategy of supplementing exogenous stem cells [33,50, valproic acid to induce differentiation of transplanted NSCs to in­
101]. terneurons. The transplant-derived interneurons were able to recon­
struct the damaged propriospinal neural network, to both receive and
send synaptic information from/to host neurons, and to directly
3.2. Exogenous neuronal relays contribute to the restoration of motor function. Lu et al. [11,14,101]
adsorbed NSCs and 10 different neurotrophic factors using fibrin
In recent study, Li et al. [60,87] developed an NT-3/fibroin-coated matrices and co-transplanted them into the SCI area, thereby greatly
gelatin sponge scaffold with sustained NT-3 release, which demon­ improving the microenvironment of the injury/graft site. Trans­
strated the ability to promote nerve fiber regeneration and to activate plantation of the NSC-derived interneurons led to remarkable axonal
endogenous stem cells to differentiate into interneurons that integrate growth and supported the formation of electrophysiological relays
into the propriospinal neural network; this scaffold has been used to across the injury/graft sites of the complete spinal cord transection,
successfully repair SCI in rats and canines. However, for complete SCI, resulting in functional recovery. However, Sharp et al. [52] and other
the insufficient quantity of endogenous stem cells may limit the effec­ researchers have demonstrated repeatedly that the direct
tiveness of this approach. Therefore, Abematsu et al. [51] applied

6
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

transplantation of human NSCs into the SCI site generates a risk of the SCI site. In such cases, the greatest advantage of the tissue engi­
abnormal proliferation and long-distance migration. Furthermore, neering strategy is its ability to meet the diverse needs of spinal cord
direct transplantation of NSCs might slows differentiation and matura­ structural and functional repair by effectively combining seed cells,
tion, resulting in delayed functional recovery of the injured spinal cord biomaterials, and bioactive factors. For example, some studies have
[102]. Lu et al. [15] implanted and assessed human H9 NSCs in an shown that stem cells can be induced to become neurons and oligo­
immunodeficient rat model of SCI over a period of 1.5 years, with the dendrocytes that can then be loaded together into a functional bioma­
grafts showing evidence of continued maturation over the entire period terial scaffold [43,100,109]. A number of studies have shown that seed
of assessment. Although biomarkers of neuronal maturity were first cells can be used as gene carriers to transplant a target gene of thera­
expressed three months after grafting, the complex processes of neuro­ peutic interest, thus achieving the dual effects of cell replacement and
genesis, neuronal pruning, and neuronal enlargement continued for one gene therapy [13,42,107,114]. Other studies have indicated that bio­
year thereafter. The authors also found that half of the axonal pro­ materials modified by bioactive factors can simultaneously be used for
jections persisted for 1.5 years, and that the number of mature oligo­ loading cells, fighting inflammation, promoting nerve regeneration, and
dendrocyte markers did not increase until one year after grafting. increasing survival of transplanted cells in SCI repair [70,75]. Therefore,
Functional recovery was first observed more than one year after the this approach not only fills the space resulting from SCI with bio­
original grafting. We analyzed a series of studies regarding the appli­ materials and cells but can also provide directional guidance to the
cation of exogenous neuronal relays as a repair strategy for SCI grafted neurons and regenerating nerve fibers [70,86].
(Table 3). Some studies have shown that, although transplanted stem Based on stem cell transplantation and tissue engineering, Zeng and
cells can survive and differentiate well into interneurons in the SCI area colleagues have developed a series of important components needed to
and even extend their long processes into the host spinal cord, the address key scientific challenges in this field, including a human NT-3
behavioral recovery of the animals is not noticeable [98,103]. Other gene recombinant adenovirus [115], a three-dimensional gelatin
studies have indicated that the key issue in the repair of motor function sponge scaffold [116], a tissue-engineered neural network [53,110],
in animals may additionally relate to whether exogenous stem scaffolds with chemotactic function [60,117], and decellularized nerve
cell-derived interneurons are able to produce the correct synaptic con­ scaffolds [99,100,118]. By combining these components with novel
nections with regenerating nerve fibers [16,31,59]. A further key point nerve tissue engineering techniques such as use of neurotrophic factors
is whether the exogenous neuronal relay can simultaneously be and their receptors, gene-modified stem cells, and biomaterials, they
replenished by a subset of oligodendrocytes to increase the remyelina­ constructed an exogenous stem cell-derived neural network tissue with
tion of regenerating nerve fibers in the injury site [59,100,104]. In functional synaptic transmission [37,110]. This tissue was then trans­
addition, another key issue that should be considered is the abnormal planted into the gap created by complete spinal cord transection
proliferation and migration of stem cells to form space-occupying lesions (removal of a 2-mm segment of rat spinal cord or a 4-mm segment of
after transplantation [52,105]. Successfully establishing an exogenous canine spinal cord). As well as improving the microenvironment at the
neuronal relay in the SCI site in a safe and effective manner remains a injury/graft site of the spinal cord, this neural network tissue was able to
challenge. promote dynamic integration and functional synergism of endogenous
stem cell-derived neurons, allowing it to function as a neuronal relay
4. Tissue-engineered neuronal relays that could repair the host propriospinal neural network (Fig. 3) [35,53,
109]. This approach avoids the uncertain differentiation of stem cells,
Research into tissue-engineered nerve regeneration and repair stra­ especially embryonic stem cells, transplanted directly into the spinal
tegies has been rapidly developing over the past decade or so, with such cord, as well as reducing the risk of abnormal proliferation and
strategies considered to offer novel treatment approaches for severe long-distance migration [52,105].
central nervous system injuries [58,110–112]. Tissue engineering is a Previous studies by Lai et al. [35,54] have shown that stem
technique that combines cell biology and materials science to construct cell-derived tissue-engineered neural network transplantation is able to
biological tissues or organs in vitro or in vivo, involving a combination act as a neuronal relay for synaptic connections between regenerating
of three elements: seed cells, biomaterials, and bioactive factors. The ascending and descending neuronal nerve fibers (Fig. 3). Marchini et al.
decision of how to assemble these three elements is based on the re­ [113] also developed an in vitro three-dimensional (3D) functional
quirements for repair of the damaged tissue or organ. Repair of tissue neuronal network derived from human neural stem cells (hNSCs). They
structure and function in a safe and effective manner is key to success­ tracked the proliferation, differentiation, and maturation of the hNSCs
fully applying tissue engineering techniques [4,58,113]. into glutamatergic, cholinergic, and GABAergic neurons in
For complete SCI, repair strategies based on tissue engineering multi-functionalized hydrogels, and evaluated how these engineered
should consider the massive loss of neurons and oligodendrocytes, neural network tissues facilitated treatment of spinal cord injuries. They
infiltration of inflammatory cells, and cavity and glial scar formation at found that transplantation of the multi-functionalized hydrogel scaffolds

Table 3
Comparison of exogenous neuronal relay strategies in different treatments of complete spinal cord transection.
Treatments Primary strategies Major advantages Major disadvantages Refs

Transplantation of NSCs or Injecting cells into the epicenter Connectivity of donor cell-derived neuronal axons, and Abnormal proliferation, uncertain [17,20,26,
NPCs of the injury site neuroprotection and immunomodulation in vivo differentiation, or long-distance 41,49,51,
migration in donor cells in vivo 52,106]
Transplantation of Seeding cells in functionalized Replacing lost neurons, repairing the propriospinal Abnormal proliferation, uncertain [11,13,15,
biomaterials loaded with materials with drug or neural network, enhancing nerve regeneration, differentiation, or long-distance 98,105,
NSCs or NPCs neurotrophic factor promoting angiogenesis, and reducing cavity size in migration in donor cells in vivo 107,108]
vivo
Transplantation of engineered Constructing stem cell-derived Replacing lost neurons and oligodendrocytes, High complexity of fabrication [19,25,37,
neural network tissue or neuronal network tissue or repairing the propriospinal neural network, enhancing technique in vitro 53–55,109]
spinal cord-like tissue spinal cord-like tissue in vitro the neuronal relay effect, and promoting remyelination
in vivo
Transplantation of fetal spinal Transplanting fetal spinal cord Replacing neurons and oligodendrocytes, repairing the Sourcing and ethical limitations in [9,10,
cord tissue tissue directly propriospinal neural network, in vivo translational medicine 56–58]

Abbreviations: NSCs, neural stem cells; NPCs, neural precursor cells.

7
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Fig. 3. Schematic diagram depicting a tissue-engineered neuronal relay. A) Fabrication process of the neural network tissue. B) Transplantation of the neural
network tissue into the injury site in completely transected spinal cord. C) Integration of the grafted neural network tissue into the host propriospinal neuronal
network, myelination of the grafted neuronal axons, and functional relay formation across the injury site in the spinal cord. SCs, Schwann cells; NSCs, neural stem
cells; 3D, three-dimensional; DRG, dorsal root ganglion; T9, 9th thoracic vertebra of the spinal cord; CST, corticospinal tract; 5-HT, 5-hydroxytryptamine; NF,
nerve fiber.

containing pre-differentiated hNSCs resulted in a greater proportion of significantly promote regeneration of brain-derived descending nerve
neuronal markers, better hNSC engraftment, and improved behavioral fibers. NSC-derived neurons were able to establish synaptic connections
recovery. Their study demonstrated that 3D cultures are better able to with descending nerve fibers, support transmission of brain-derived
mimic the conditions to which cells are normally exposed, and further excitatory neural information, and promote improvement of motor
showed that neural network tissue formed by the pre-differentiated function. Thus, SCLT transplantation shows promise for application to
hNSCs in vitro is able to integrate and form functional circuits in the repair of complete SCI.
rat spinal cord within 6 weeks. However, this technique requires further It has been acknowledged that research on complete SCI repair
development as functional integration of tissue-engineered neural should not only focus on the effects of spinal nerve fiber regeneration
network tissue into the host propriospinal neural network remains and the transmission of ascending and descending neural information
limited, which in turn limits improvement of autonomous motor func­ but also consider disuse atrophy of neurons and muscles deprived of
tion in experimental animals. Recent literature suggests that disordered functional innervation [67,93,119]. The current work of Lai et al. [54]
regeneration of nerve fibers or disordered distribution of grafted neu­ shows that transplanted neural network tissue or SCLT can induce
rons is not conducive to repair of spinal cord structure and function brain-derived nerve fiber regeneration in the injury/graft area of a
[100]. In order to improve the efficiency of functional integration be­ complete spinal cord transection, achieving synaptic contacts with
tween grafted and host neurons, it will be necessary to develop a new transplanted stem cell-derived neurons to enable functional integration.
scaffold that can guide directional axon regeneration and the distribu­ However, adverse effects can occur before functional integration of the
tion of transplanted cells [70,99]. neural network tissue or the SCLT into the host spinal cord is complete.
Based on the hypothesis that a tissue-engineered neuronal relay can Specifically, pathophysiological changes in the injured spinal cord can
be used to repair spinal cord defects, and given the progress made in result in functional silencing or disuse atrophy of the propriospinal
stem cell tissue engineering and tissue organ construction techniques, neurons at the injury/graft site, the caudal spinal cord, and the para­
Lai et al. [54] combined NSCs, neurotrophic factors and their receptors, lyzed limb muscles, which may limit repair of spinal cord defects [30,
and a collagen scaffold to construct a spinal cord-like tissue (SCLT) in 40]. Therefore, rehabilitation physiotherapy (for example functional
vitro for the first time. This SCLT is similar to normal white matter and electrical stimulation) should be used in combination with the stem
gray matter of the spinal cord and has architectural, phenotypic, and cell-derived neuronal relay strategy to restore innervation of spinal cord
functional similarities to adult rat spinal cord. It contains the corre­ motor neurons and their targeted muscle cells as early as possible [1,5,
sponding major cell types (neurons, oligodendrocytes, and astrocytes), 120]. In the anterograde direction, a variety of factors (including
and is composed of white matter-like and gray matter-like tissue mod­ acetylcholine and neurotrophic factors) secreted by spinal motor neu­
ules. This modular construction of the SCLT offers the possibility of rons are transported through motor nerve fibers to maintain the struc­
structural and functional repair after complete spinal cord transection. ture and function of target muscle cells, and to prevent muscle atrophy
Transplantation of this SCLT into the gap of a complete spinal cord [119,120]. In the retrograde direction, myogenic trophic factors
transection (with a 2-mm-long defective tissue area) was able to secreted by muscle cells are transported via the motor nerve fibers to the

8
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

spinal cord to maintain structure and function of the target motor neu­ promoting weight-bearing locomotion of paralyzed legs in monkeys
rons, and to prevent neuronal atrophy [121]. [122].
The Lancet Neurology also reviewed the use of a brain–spine interface
5. Brain-spine interfaces and electrical stimulation to record and decode brain signals to restore reaching and grasping
movements via brain-controlled spinal cord stimulation [61]. These
Based on the observation that brain-derived descending nerve fibers studies revealed that repeated spinal cord electrical stimulation and
such as the CST have difficulty in regenerating across a complete spinal rehabilitation training could enhance autonomous movement [121,
cord transection to reconnect with target neurons, Courtine and col­ 124–126]. Harkema et al. [38] described a 23 year-old man who was
leagues developed a brain–spine interface which could bypass the lesion paraplegic due to a C7-T1 subluxation, with complete loss of clinically
[122,123]. This strategy was able to restore control over leg muscle detectable voluntary motor function with partial preservation of
activity for walking using epidural electrical stimulation (EES; Fig. 4). sensation below the T1 cord segment. The authors placed a 16-electrode
Researchers interfaced motor cortex activity guiding leg movement with array on the dura to allow for chronic electrical stimulation. They sub­
EES protocols to establish a brain-spine interface that alleviated gait sequently found that the patient was able to produce standing- and
deficits in a rhesus monkey with SCI. An intracortical microelectrode locomotor-like patterns during stimulation using standing-specific pa­
array was implanted into the region of the motor cortex in the brain that rameters and bilateral load-bearing proprioceptive input. Further
guides leg movement, and a spinal cord electrical stimulation system research has confirmed that EES is able to modulate the propriospinal
composed of a pulse generator and a spatially selective epidural implant neural network in humans into a physiological state that enables sensory
was placed in the epidural area of the spinal cord. The design and input from standing and stepping movements to serve as a source of
implementation of the control system linked online neural decoding of neural control [22,127,128]. The above studies indicate that a brain–­
flexion and extension motor states with electrical stimulation protocols, spine interface has the potential to serve as an “artificial intelligence

Fig. 4. Schematic diagram showing two approaches


to spinal cord stimulation in the brain–spine inter­
face. Intracortical microelectrode array are implanted
in the motor cortex of the cerebrum to record motor
information in the cerebral cortex; this information is
used to form the input stimulation signals to the
spinal cord, using computer-based decoding. During
this process, a pulse generator is required to generate
a connection with the input stimulation signal elec­
trodes in the spinal cord. EES: The input stimulation
signal electrodes are placed on the dural surface of
the spinal cord, in order to affect the afferent
neuronal circuits and the intrinsic interneuronal cir­
cuit in the spinal cord. ISMS: The input stimulation
signal micro-needle electrodes are accurately inserted
into the spinal cord tissue to transmit stimulation
signals to the intrinsic interneuronal circuits in the
spinal cord. EES, epidural electrical stimulation;
ISMS, intraspinal microstimulation; SCI, spinal cord
injury; DRG, dorsal root ganglion.

9
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

relay” where brain-derived neuronal signals are first converted into an 6. Combinatorial treatments using electrical stimulation and
electrical signal and then reconverted into neuronal signals in the tissue-engineered neuronal relays
injured spinal cord (Fig. 4).
Another spinal cord electrical stimulation approach that is able to Recent studies in brain–spine interfaces and electrical stimulation
restore motor function is intraspinal microstimulation (ISMS) [129, have suggested that EES or ISMS are able to stimulate functionally silent
130]. ISMS differs from EES in that it delivers the electrical signal via (inactivated) descending neural pathways in injured spinal cord [129,
microelectrodes implanted within the spinal cord [131]. To date, the use 130,140]. Brain-spinal interfaces and electrical stimulation, by
of ISMS strategies is rare in humans, but animal experiments have enhancing electrical activity of cortex pyramidal neurons, can promote
provided insights into the potential benefits of this approach [132,133]. regeneration of axons in the CST of a damaged spinal cord. These re­
In animal models, ISMS can elicit a wide variety of function-related generated CST fibers expressing protein tyrosine phosphatase-σ (PTPσ)
movements, such as grasping, reaching, and stepping [134–136]. may specifically bind to NSC-derived neurons overexpressing NT-3 re­
When ISMS microelectrodes were implanted into the ventral spinal cord, ceptor tyrosine kinase C (TrkC) at the injury/graft site of a spinal cord,
direct activation of ventral root nerve fibers or motor neurons was enhancing the possibility of forming synaptic structures between them.
observed, leading to single joint movement [129,137]. In contrast, when PTPσ in the presynaptic membrane binds to TrkC in the postsynaptic
the microelectrodes were implanted into the intermediate lamina of the membrane, thereby enhancing adhesion of the presynaptic membrane
spinal cord, the electrical stimulation was most likely to subsequently and promoting development of excitatory synapses [152]. NT-3 can
activate interneurons and nerve fibers [130,138–140]. The interneurons further promote adhesion between PTPσ and TrkC [153]. It was
in turn activated a complex propriospinal neural network, resulting in observed that a small number of CST fibers can regenerate into the
coordinated motion. Paired with the brain–spine interface system, ISMS injury/graft site to connect with stem cell-derived neurons over­
may be especially useful for controlling the electrical current based on expressing TrkC in response to an exogenous NT-3 concentration
activity-related signals from the brain that occur when an animal at­ gradient [19,25,154]. Furthermore, electrical stimulation might also
tempts to move (Fig. 4) [134,141,142]. This kind of activity-dependent provoke relay neurons at the injury site that innervate the caudal spinal
electrical stimulation might also be used in SCI rehabilitation in the cord to transmit their signals to target muscle cells, enabling standing
future to strengthen cortico-spinal connections and to regulate synaptic and walking movements [127,155,156].
plasticity in the propriospinal neural network [124,143]. Many studies Transcutaneous spinal cord stimulation (tcSCS) is a noninvasive
suggest that EES or ISMS may be useful in directing the rehabilitation of method in which electrodes are placed on the surface of the skin above
specific motor pathways. Research has established that the spinal cord the spinal column and modulated intermediate-frequency currents
undergoes neural remodeling after injury, and that this process of (medium frequency electrical stimulation) are used to activate dorsal
remodeling can lead to synaptic plasticity in neural pathways, thus root ganglion neurons and propriospinal neurons; this method is also
inducing beneficial motor effects [127,143]. Thus, targeted therapeutic used for functional repair following SCI (Fig. 5) [157–159]. Medium
EES or ISMS may help to guide these remodeling mechanisms towards frequency electrical stimulation does not generate tissue electrolysis.
the formation of functional motor pathways, particularly when the Furthermore, it can overcome the electrical resistance of body tissues,
neuromuscular system has not yet undergone atrophy following chronic penetrate deeply into spinal tissue, and increase the excitability of
paralysis. The observed improvements in muscle mass and other phys­ propriospinal neurons and muscles without exciting afferent nerve fibers
iological functions support the notion that EES or ISMS can help to involved in algesthesia [160]. Emerging evidence suggests that EES and
counteract such deterioration [144–147]. However, there are still many tcSCS activate the same propriospinal neural network, and that both can
challenges, as the limitations of microelectrode implantation at this regulate the synaptic plasticity of neural networks, promote neuro­
stage mean that it is only possible to record a small amount of brain trophic factor secretion, and improve the spinal cord microenvironment
activity and there is a risk of infection. Moreover, the microelectrode [159,161]. Although early results from functional electrical stimulation
implanted into brain or spinal cord can cause local immune responses, of the spinal cord indicate that it can play a significant role in promoting
induce the generation of glial scars around the electrode, and result in motor recovery, tapping its full potential to elicit sustained, long-term
the reduced efficacy of the brain–spine interface system [148]. In improvements will likely require a combination of treatments [22,162].
addition, the implanted microelectrode also has limited longevity and Rodent studies have demonstrated that functional electrical stimu­
stability, with failure typically occurring approximately one year lation may be particularly useful when combined with physical reha­
post-implantation [149]. Although the brain–spine interface can suc­ bilitation to enable activation of specific motor pathways or
cessfully transmit brain-derived motor nerve signals to the spinal cord, strengthening of synaptic connections between neurons. Even weeks
the first generation of such artificial intelligence relay technology sys­ after electrical stimulation has ceased, treated animals show lasting
tems cannot return the sensory nerve signals of the paralyzed patients to motor improvements [63,120,163]. These results suggest long-term
the brain [121,150]. Therefore, there is still a crucial role for biomimetic therapeutic effects of electrical stimulation of the descending path­
medical techniques in repairing SCI at the structural and functional ways. These functional electrical stimulation technologies might also be
levels. combined with tissue-engineered neuronal relay strategies, stem cell
Increasing numbers of studies have suggested that more effective SCI transplantation, pharmacological interventions, and training activities
repair can be achieved by a combination of multiple strategies, such as [22,91,154].
cell and tissue transplantation, application of neurotrophic factors for The results Jin and his colleagues obtained for repair of SCI using
axon and myelin regeneration, delivery of neuroprotective drugs or spinal cord electrical stimulation in combination with a tissue-
immunosuppressants, and regulation of neural circuit plasticity by engineered neuronal relay suggest that electrical stimulation of the
physical therapy and rehabilitation training [125,151]. Optimal com­ tail nerve or electroacupuncture (EA) can be used to upregulate
binations of the above treatment strategies will provide inspiration for expression of endogenous NT-3 in the spinal cord after its complete
improved clinical treatments. We suggest that combinatorial treatments transection [55,67]. Tail nerve electrical stimulation falls into the
using physical therapy and tissue-engineering neuronal relay could category of tcSCS methods, while EA refers to the application of a pulsed
regulate synaptic plasticity and promote effective integration of current to the needle and is based on traditional Chinese acupuncture
neuronal relays and the propriospinal neural network. (acupoint treatment with a needle (Fig. 5) [164]). EA can maintain a
continuous state of stimulation in the body for long periods due to the
physiological effect of pulsed electricity, thereby providing the dual
benefits of acupoint and electrical stimulation [112,165]. The mecha­
nism of its therapeutic effect on SCI mainly involves improvement of the

10
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Fig. 5. Schematic diagram showing two methods of body surface electrical stimulation. EA: A pulse generator (a medical EA apparatus) generates the pulse current.
This is transmitted through the acupuncture needle at the acupoint to stimulate sensory nerve endings of the meningeal branch of the spinal nerve, affecting the
afferent neuronal circuit in the spinal cord. tcSCS: An electrode patch is placed on the surface of the skin above the spinal column, and then a pulse generator is used
to generate a modulated intermediate-frequency current to activate DRG and propriospinal neurons. EA, electroacupuncture; tcSCS, transcutaneous spinal cord
stimulation; DRG, dorsal root ganglion.

microenvironment of the injured area, reducing secondary injury, spinal cord via stimulating the nerve endings of meningeal branch
reducing scar tissue formation, and promoting nerve regeneration (containing the sensory afferent fibers from the dorsal root ganglion),
[166]. Several studies have reported that EA treatment can lead to acting on CGRP receptors in the spinal cord neurons. CGRP excites
obvious improvements in hindlimb locomotor and sensory function in L-type voltage-gated Ca2+ channels (VGCCs) on the cell membrane by
SCI rats, and have concluded that EA may promote recovery of neuro­ up-regulating the calcium/calmodulin-dependent protein kinase
plasticity via systematic regulation of neurotrophic factors and their (αCaMKII) pathway, promotes differentiation of neurons, and stimulates
receptors [44,167,168]. Due to the action of the pulsed electric fields, a secretion of NT-3 [165]. If tissue-engineered neural network tissue is
large number of residual propriospinal neurons and ascending and transplanted and then stimulated by EA, it is possible to promote
descending nerve fibers are also stimulated, overcoming inhibition due continuous synthesis and secretion of endogenous NT-3 in the injured
to inactivity and helping to restore normal function. Neurons that are spinal cord so as to enable targeted growth of the axons of stem
partially damaged are able to recover, restoring functional connections cell-derived neurons, establish synaptic connections with host neurons
with the propriospinal neural network (Fig. 5) [55,154,165]. of the spinal cord, and improve limb motor function. In addition, EA
Following the transplantation of NSC-derived neural network tissues stimulation could increase the level of NT-3 in the rostral and caudal
into the gap resulting from complete spinal cord transection, Jin et al. areas adjacent to the injured spinal cord, and the concentration of NT-3
[55] showed that use of EA to regulate the transplanted area and its in both areas was higher than in the injured area itself. Therefore, gra­
adjacent tissue microenvironment was able to achieve the following: dients of NT-3 concentration are formed between the rostral/caudal
promotion of cell synthesis and secretion of NT-3, activation of the areas and the injured area, which facilitates axon-targeting growth of
downstream TrkC/AKT/mTOR (TrkC, NT-3 receptor, AKT, AKT kinase, transplanted mesenchymal stem cell-derived neuron-like cells over­
mTOR, mammalian target of rapamycin) signaling pathway in target expressing TrkC, enhance synaptic connections with host spinal neurons
cells, increase of survival and neuronal differentiation in transplanted secreting endogenous NT-3, and promote integration of the transplanted
neural network tissues, maintenance of synaptic connections, and stem cell-derived neural network with host neural circuits in the injured
enhanced functional integration of tissue with the host spinal neural spinal cord [55,112].
network. EA in Governor vessel acupoints may increases release of Although the results obtained from functional electrical stimulation
calcitonin gene-related peptide (CGRP) in afferent nerve endings in the of the spinal cord at the locations described above for EES, ISMS, tcSCS,

11
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

and EA has led to substantial progress in the field (Fig. 6; Table 4), we transplanted tissue-engineered neural network tissue from a disordered
have limited insight into the underlying mechanisms responsible for the to an ordered state. Concurrent limb movement training should also help
functional responses of the spinal cord to electrical stimulation. Never­ to accelerate ordering of tissue-engineered neural network tissue
theless, electrical stimulation does improve the microenvironment of the (Fig. 6), which can then better act as a neuronal relay.
SCI area, activates the propriospinal neuronal network, regulates syn­
aptic remodeling, and increases motor function in paralyzed patients 7. Conclusion and future perspectives
[67,169–171]. In future studies, continuous use of neuroregulatory
techniques in the cerebral cortex, including transcranial magnetic In summary, post-SCI damage can be repaired by both endogenous
stimulation (TMS), brain-spinal interface stimulation, photogenetic and exogenous neuronal relay strategies. Endogenous neuronal relay
stimulation, and chemogenetic stimulation may help to transform strategies include: (1) compensatory neuronal relays (which take

Fig. 6. Schematic diagram showing the combined strategies using a tissue-engineered neuronal relay and electrical stimulation. Approaches are grouped by the type
of spinal cord electrical stimulation: A) EA treatment; B) tcSCS; C) EES; D) ISMS; E) Summary of the electrical stimulation pathway. tcSCS, transcutaneous spinal cord
stimulation; EA, electroacupuncture; EES, epidural electrical stimulation; ISMS, intraspinal microstimulation; DRG, dorsal root ganglion; CST, corticospinal tract; 5-
HT, 5-hydroxytryptamine; TH, tyrosine hydroxylase; NFs, nerve fibers.

12
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

Table 4
Treatment protocols of functional electrical stimulation strategies for spinal cord injury SCI.
Functional Treatment protocols Key outcomes Major advantages Major disadvantages Refs
electrical
stimulation

Epidural Surgical implantation of Enabling voluntary control of Modulates the propriospinal Invasive; facilitates locomotion only [122–125,
electrical microelectrode array into the walking and sustaining active neural network, activates via a narrow range of stimulation 150,156,172,
stimulation epidural surface of spinal movement silent neuronal circuits, and parameters 173]
(EES) cord promotes synaptic plasticity
Intraspinal Stimulating the propriospinal Promoting and sustaining Accurately targets specific Invasive; causes gliosis and electrode [129,130,
microstimu- locomotor neural network motor recovery, and enabling spinal locomotor circuitry damage, and eventually results in 132,133]
lation (ISMS) via an intraspinal long distance walking inefficient stimulation
microelectrode array
Transcutan-eous Placing electrodes on the skin Modulating the excitability of Enables noninvasive Targets a specific area of spinal cord [38,157,158,
spinal cord surface of the vertebral the propriospinal neural treatment, with a convenient less accurately compared with EES and 161,174]
stimulation column network and evoking rhythmic and inexpensive commercial ISMS
(tcSCS) limb movement stimulation device
Electroacu- Application of pulsed current Effectively increasing Achieves double efficacy with Targets a specific area of spinal cord [154,164,
puncture (EA) to acupuncture needles neurotrophic factor secretion, combined acupoint and less accurately compared with EES and 166,170,
inhibiting inflammation, and electrical stimulation; ISMS; mostly used in analgesia and 175]
promoting nerve regeneration minimally invasive less so for the recovery of locomotor
function

advantage of the intact propriospinal neuronal axons in the spinal cord Guangzhou City (201704020221), the Foundation of Guangdong Prov­
using the collateral sprouting method to repair the damaged neural ince (2017B020210012) and the 111 Project for Academic Exchange
network), and (2) autologous stem cell-derived neuronal relays (in Program (B13037) to Y. S. Zeng and X. Zeng.
which a neural network is formed by the differentiation of spinal cord
stem cells). Exogenous neuronal relays include the following: (1) References
transplanted embryonic spinal cord tissue-derived neuronal relays
(which used embryonic spinal cord tissue to repair the damaged host [1] A.E. Ropper, A.H. Ropper, Acute spinal cord compression, N. Engl. J. Med. 376
(14) (2017) 1358–1369.
neural network), (2) transplanted stem cell-derived neuronal relays [2] A.E. Ropper, D.K. Thakor, I. Han, D. Yu, X. Zeng, J.E. Anderson, Z. Aljuboori, S.
(which use differentiation of transplanted stem cells in the SCI area to W. Kim, H. Wang, R.L. Sidman, R.D. Zafonte, Y.D. Teng, Defining recovery
form a neural network), (3) tissue-engineered neuronal relays (Fig. 3) neurobiology of injured spinal cord by synthetic matrix-assisted hMSC
implantation, Proc. Natl. Acad. Sci. U S A. 114 (5) (2017) E820–E829.
which involve the application of tissue-engineered neural network tis­ [3] L.M. Ramer, M.S. Ramer, E.J. Bradbury, Restoring function after spinal cord
sues or SCLTs constructed in vitro), and (4) artificial intelligence relays injury: towards clinical translation of experimental strategies, Lancet Neurol. 13
(i.e., brain–spine interfaces). Regardless of which neuronal relay strat­ (12) (2014) 1241–1256.
[4] M.A. Anderson, T.M. O’Shea, J.E. Burda, Y. Ao, S.L. Barlatey, A.M. Bernstein, J.
egy is adopted for SCI repair, the key question is how to better achieve H. Kim, N.D. James, A. Rogers, B. Kato, A.L. Wollenberg, R. Kawaguchi,
functional integration with the propriospinal neural network; this re­ G. Coppola, C. Wang, T.J. Deming, Z. He, G. Courtine, M.V. Sofroniew, Required
mains to be investigated in future studies [16,18,102,176]. In the future, growth facilitators propel axon regeneration across complete spinal cord injury,
Nature 561 (7723) (2018) 396–400.
we envisage that for complete spinal cord transection or for larger spinal
[5] J. Donovan, S. Kirshblum, Clinical trials in traumatic spinal cord injury,
cord defects, biomimetic medical techniques will be available to provide Neurotherapeutics : J. Am. Soc. Exp. NeuroTher. 15 (3) (2018) 654–668.
tissue-engineered neuronal relays for the timely re-construction of [6] A. Blesch, M.H. Tuszynski, Spinal cord injury: plasticity, regeneration and the
neural networks in the spinal cord. The combination of such techniques challenge of translational drug development, Trends Neurosci. 32 (1) (2009)
41–47.
with electrical stimulation of propriospinal neurons could enable acti­ [7] K. Liu, Y. Lu, J.K. Lee, R. Samara, R. Willenberg, I. Sears-Kraxberger, A. Tedeschi,
vation of neuronal circuits that are functionally silent, owing to the loss K.K. Park, D. Jin, B. Cai, B. Xu, L. Connolly, O. Steward, B. Zheng, Z. He, PTEN
of brain-derived neural information (Fig. 6). Electrical stimulation deletion enhances the regenerative ability of adult corticospinal neurons, Nat.
Neurosci. 13 (9) (2010) 1075–1081.
should enable regulation of synaptic plasticity in the propriospinal [8] S. Hou, Relay strategies combined with axon regeneration: a promising approach
neural network, and furthermore help stem cell-derived neurons from to restore spinal cord injury, Neural regen. res. 9 (12) (2014) 1177–1179.
the tissue-engineered neural network tissue or SCLT to establish more [9] P.J. Reier, B.S. Bregman, J.R. Wujek, Intraspinal transplantation of embryonic
spinal cord tissue in neonatal and adult rats, J. Comp. Neurol. 247 (3) (1986)
accurate synaptic connections with the host propriospinal neurons, 275–296.
achieve functional integration, and repair autonomous motor function [10] J.D. Houle, P.J. Reier, Transplantation of fetal spinal cord tissue into the
in paralyzed patients (Fig. 6). chronically injured adult rat spinal cord, J. Comp. Neurol. 269 (4) (1988)
535–547.
[11] P. Lu, Y. Wang, L. Graham, K. McHale, M. Gao, D. Wu, J. Brock, A. Blesch, E.
Declaration of competing interest S. Rosenzweig, L.A. Havton, B. Zheng, J.M. Conner, M. Marsala, M.H. Tuszynski,
Long-distance growth and connectivity of neural stem cells after severe spinal
cord injury, Cell 150 (6) (2012) 1264–1273.
The authors declare that they have no known competing financial [12] X. Li, D. Liu, Z. Xiao, Y. Zhao, S. Han, B. Chen, J. Dai, Scaffold-facilitated
interests or personal relationships that could have appeared to influence locomotor improvement post complete spinal cord injury: motor axon
the work reported in this paper. regeneration versus endogenous neuronal relay formation, Biomaterials 197
(2019) 20–31.
[13] X. Zhang, Y. Zeng, W. Zhang, J. Wang, J. Wu, J. Li, Co-transplantation of neural
Acknowledgements stem cells and NT-3-overexpressing Schwann cells in transected spinal cord,
J. Neurotrauma 24 (12) (2007) 1863–1877.
[14] P. Lu, K. Kadoya, M.H. Tuszynski, Axonal growth and connectivity from neural
This work was supported by grants from the Chinese National Nat­ stem cell grafts in models of spinal cord injury, Curr. Opin. Neurobiol. 27 (2014)
ural Science Foundation (81891003, 8167150879) to Y. S. Zeng; from 103–109.
the National Key R&D Program of China (2017YFA0104700), the Young [15] P. Lu, S. Ceto, Y. Wang, L. Graham, D. Wu, H. Kumamaru, E. Staufenberg, M.
H. Tuszynski, Prolonged human neural stem cell maturation supports recovery in
Elite Scientist Sponsorship Program by CAST (YESS, 2018QNRC001), injured rodent CNS, J. Clin. Investig. 127 (9) (2017) 3287–3299.
the Fundamental Research Funds for the Central Universities [16] J.F. Bonner, O. Steward, Repair of spinal cord injury with neuronal relays: from
(18ykpy38), and the Chinese National Natural Science Foundation fetal grafts to neural stem cells, Brain Res. 1619 (2015) 115–123.
(81971157) to B. Q. Lai; and from the Co-innovation Foundation of

13
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

[17] P. Assinck, G.J. Duncan, B.J. Hilton, J.R. Plemel, W. Tetzlaff, Cell transplantation [41] E.S. Rosenzweig, J.H. Brock, P. Lu, H. Kumamaru, E.A. Salegio, K. Kadoya, J.
therapy for spinal cord injury, Nat. Neurosci. 20 (5) (2017) 637–647. L. Weber, J.J. Liang, R. Moseanko, S. Hawbecker, J.R. Huie, L.A. Havton, Y.
[18] P. Lu, Stem cell transplantation for spinal cord injury repair, Prog. Brain Res. 231 S. Nout-Lomas, A.R. Ferguson, M.S. Beattie, J.C. Bresnahan, M.H. Tuszynski,
(2017) 1–32. Restorative effects of human neural stem cell grafts on the primate spinal cord,
[19] G.H. Wu, H.J. Shi, M.T. Che, M.Y. Huang, Q.S. Wei, B. Feng, Y.H. Ma, L.J. Wang, Nat. Med. 24 (4) (2018) 484–490.
B. Jiang, Y.Q. Wang, I. Han, E.A. Ling, X. Zeng, Y.S. Zeng, Recovery of paralyzed [42] Q. Cao, Q. He, Y. Wang, X. Cheng, R.M. Howard, Y. Zhang, W.H. DeVries, C.
limb motor function in canine with complete spinal cord injury following B. Shields, D.S. Magnuson, X.M. Xu, D.H. Kim, S.R. Whittemore, Transplantation
implantation of MSC-derived neural network tissue, Biomaterials 181 (2018) of ciliary neurotrophic factor-expressing adult oligodendrocyte precursor cells
15–34. promotes remyelination and functional recovery after spinal cord injury,
[20] S. Nishimura, A. Yasuda, H. Iwai, M. Takano, Y. Kobayashi, S. Nori, O. Tsuji, J. Neurosci. : off. J. Soc. Neurosci. 30 (8) (2010) 2989–3001.
K. Fujiyoshi, H. Ebise, Y. Toyama, H. Okano, M. Nakamura, Time-dependent [43] S. Erceg, M. Ronaghi, M. Oria, M.G. Rosello, M.A. Arago, M.G. Lopez,
changes in the microenvironment of injured spinal cord affects the therapeutic I. Radojevic, V. Moreno-Manzano, F.J. Rodriguez-Jimenez, S.S. Bhattacharya,
potential of neural stem cell transplantation for spinal cord injury, Mol. Brain 6 J. Cordoba, M. Stojkovic, Transplanted oligodendrocytes and motoneuron
(2013) 3. progenitors generated from human embryonic stem cells promote locomotor
[21] Y. Zheng, Y.R. Mao, T.F. Yuan, D.S. Xu, L.M. Cheng, Multimodal treatment for recovery after spinal cord transection, Stem Cell. 28 (9) (2010) 1541–1549.
spinal cord injury: a sword of neuroregeneration upon neuromodulation, Neural [44] Z. Yang, A. Zhang, H. Duan, S. Zhang, P. Hao, K. Ye, Y.E. Sun, X. Li, NT3-chitosan
regen. res. 15 (8) (2020) 1437–1450. elicits robust endogenous neurogenesis to enable functional recovery after spinal
[22] M. Bonizzato, N.D. James, G. Pidpruzhnykova, N. Pavlova, P. Shkorbatova, cord injury, Proc. Natl. Acad. Sci. U S A. 112 (43) (2015) 13354–13359.
L. Baud, C. Martinez-Gonzalez, J.W. Squair, J. DiGiovanna, Q. Barraud, S. Micera, [45] J.S. Rao, C. Zhao, A. Zhang, H. Duan, P. Hao, R.H. Wei, J. Shang, W. Zhao, Z. Liu,
G. Courtine, Multi-pronged neuromodulation intervention engages the residual J. Yu, K.S. Fan, Z. Tian, Q. He, W. Song, Z. Yang, Y.E. Sun, X. Li, NT3-chitosan
motor circuitry to facilitate walking in a rat model of spinal cord injury, Nat. enables de novo regeneration and functional recovery in monkeys after spinal
Commun. 12 (1) (2021) 1925. cord injury, Proc. Natl. Acad. Sci. U S A. 115 (24) (2018) E5595–E5604.
[23] S. Pan, Z. Qi, Q. Li, Y. Ma, C. Fu, S. Zheng, W. Kong, Q. Liu, X. Yang, Graphene [46] Z. Su, W. Niu, M.L. Liu, Y. Zou, C.L. Zhang, In vivo conversion of astrocytes to
oxide-PLGA hybrid nanofibres for the local delivery of IGF-1 and BDNF in spinal neurons in the injured adult spinal cord, Nat. Commun. 5 (2014) 3338.
cord repair, Artificial cells, Nanomed Biotechnol 47 (1) (2019) 651–664. [47] L.L. Wang, C.L. Zhang, Engineering new neurons: in vivo reprogramming in
[24] H. Zhang, X. Fang, D. Huang, Q. Luo, M. Zheng, K. Wang, L. Cao, Z. Yin, mammalian brain and spinal cord, Cell Tissue Res. 371 (1) (2018) 201–212.
Erythropoietin signaling increases neurogenesis and oligodendrogenesis of [48] M. Zarei-Kheirabadi, M. Hesaraki, S. Kiani, H. Baharvand, In vivo conversion of
endogenous neural stem cells following spinal cord injury both in vivo and in rat astrocytes into neuronal cells through neural stem cells in injured spinal cord
vitro, Mol. Med. Rep. 17 (1) (2018) 264–272. with a single zinc-finger transcription factor, Stem Cell Res. Ther. 10 (1) (2019)
[25] X. Zeng, X.C. Qiu, Y.H. Ma, J.J. Duan, Y.F. Chen, H.Y. Gu, J.M. Wang, E.A. Ling, J. 380.
L. Wu, W. Wu, Y.S. Zeng, Integration of donor mesenchymal stem cell-derived [49] Y. Ogawa, K. Sawamoto, T. Miyata, S. Miyao, M. Watanabe, M. Nakamura, B.
neuron-like cells into host neural network after rat spinal cord transection, S. Bregman, M. Koike, Y. Uchiyama, Y. Toyama, H. Okano, Transplantation of in
Biomaterials 53 (2015) 184–201. vitro-expanded fetal neural progenitor cells results in neurogenesis and functional
[26] M.H. Tuszynski, O. Steward, Concepts and methods for the study of axonal recovery after spinal cord contusion injury in adult rats, J. Neurosci. Res. 69 (6)
regeneration in the CNS, Neuron 74 (5) (2012) 777–791. (2002) 925–933.
[27] S. Quraishe, L.H. Forbes, M.R. Andrews, The Extracellular Environment of the [50] H.E. Olson, G.E. Rooney, L. Gross, J.J. Nesbitt, K.E. Galvin, A. Knight, B. Chen, M.
CNS: Influence on Plasticity, Sprouting, and Axonal Regeneration after Spinal J. Yaszemski, A.J. Windebank, Neural stem cell- and Schwann cell-loaded
Cord Injury, Neural Plasticity 2018, 2018, p. 2952386. biodegradable polymer scaffolds support axonal regeneration in the transected
[28] H. Liu, X. Xu, Y. Tu, K. Chen, L. Song, J. Zhai, S. Chen, L. Rong, L. Zhou, W. Wu, K. spinal cord, Tissue engineering, Part. Accel. 15 (7) (2009) 1797–1805.
F. So, S. Ramakrishna, L. He, Engineering microenvironment for endogenous [51] M. Abematsu, K. Tsujimura, M. Yamano, M. Saito, K. Kohno, J. Kohyama,
neural regeneration after spinal cord injury by reassembling extracellular matrix, M. Namihira, S. Komiya, K. Nakashima, Neurons derived from transplanted
ACS Appl. Mater. Interfaces 12 (15) (2020) 17207–17219. neural stem cells restore disrupted neuronal circuitry in a mouse model of spinal
[29] D. Ma, Y. Zhao, L. Huang, Z. Xiao, B. Chen, Y. Shi, H. Shen, J. Dai, A novel cord injury, J. Clin. Investig. 120 (9) (2010) 3255–3266.
hydrogel-based treatment for complete transection spinal cord injury repair is [52] K.G. Sharp, K.M. Yee, O. Steward, A re-assessment of long distance growth and
driven by microglia/macrophages repopulation, Biomaterials 237 (2020) connectivity of neural stem cells after severe spinal cord injury, Exp. Neurol. 257
119830. (2014) 186–204.
[30] B. Chen, Y. Li, B. Yu, Z. Zhang, B. Brommer, P.R. Williams, Y. Liu, S.V. Hegarty, [53] B.Q. Lai, J.M. Wang, J.J. Duan, Y.F. Chen, H.Y. Gu, E.A. Ling, J.L. Wu, Y.S. Zeng,
S. Zhou, J. Zhu, H. Guo, Y. Lu, Y. Zhang, X. Gu, Z. He, Reactivation of dormant The integration of NSC-derived and host neural networks after rat spinal cord
relay pathways in injured spinal cord by KCC2 manipulations, Cell 174 (3) (2018) transection, Biomaterials 34 (12) (2013) 2888–2901.
521–535 e13. [54] B.Q. Lai, B. Feng, M.T. Che, L.J. Wang, S. Cai, M.Y. Huang, H.Y. Gu, B. Jiang, E.
[31] L.T. Alto, L.A. Havton, J.M. Conner, E.R. Hollis 2nd, A. Blesch, M.H. Tuszynski, A. Ling, M. Li, X. Zeng, Y.S. Zeng, A modular assembly of spinal cord-like tissue
Chemotropic guidance facilitates axonal regeneration and synapse formation allows targeted tissue repair in the transected spinal cord, Adv Sci (Weinh.Baden-
after spinal cord injury, Nat. Neurosci. 12 (9) (2009) 1106–1113. Wurttemberg, Germany) 5 (9) (2018) 1800261.
[32] M.V. Sofroniew, Dissecting spinal cord regeneration, Nature 557 (7705) (2018) [55] H. Jin, Y.T. Zhang, Y. Yang, L.Y. Wen, J.H. Wang, H.Y. Xu, B.Q. Lai, B. Feng, M.
343–350. T. Che, X.C. Qiu, Z.L. Li, L.J. Wang, J.W. Ruan, B. Jiang, X. Zeng, Q.W. Deng,
[33] K. Kadoya, P. Lu, K. Nguyen, C. Lee-Kubli, H. Kumamaru, L. Yao, J. Knackert, G. Li, Y. Ding, Y.S. Zeng, Electroacupuncture facilitates the integration of neural
G. Poplawski, J.N. Dulin, H. Strobl, Y. Takashima, J. Biane, J. Conner, S.C. Zhang, stem cell-derived neural network with transected rat spinal cord, Stem cell
M.H. Tuszynski, Spinal cord reconstitution with homologous neural grafts enables reports 12 (2) (2019) 274–289.
robust corticospinal regeneration, Nat. Med. 22 (5) (2016) 479–487. [56] J.D. Houle, P.J. Reier, Regrowth of calcitonin gene-related peptide (CGRP)
[34] Y. Ohori, S. Yamamoto, M. Nagao, M. Sugimori, N. Yamamoto, K. Nakamura, immunoreactive axons from the chronically injured rat spinal cord into fetal
M. Nakafuku, Growth factor treatment and genetic manipulation stimulate spinal cord tissue transplants, Neurosci. Lett. 103 (3) (1989) 253–258.
neurogenesis and oligodendrogenesis by endogenous neural progenitors in the [57] Y. Iwashita, S. Kawaguchi, M. Murata, Restoration of function by replacement of
injured adult spinal cord, J. Neurosci. : off. J. Soc. Neurosci. 26 (46) (2006) spinal cord segments in the rat, Nature 367 (6459) (1994) 167–170.
11948–11960. [58] J. Ganz, E. Shor, S. Guo, A. Sheinin, I. Arie, I. Michaelevski, S. Pitaru, D. Offen,
[35] B.Q. Lai, M.T. Che, B. Feng, Y.R. Bai, G. Li, Y.H. Ma, L.J. Wang, M.Y. Huang, Y. S. Levenberg, Implantation of 3D constructs embedded with oral mucosa-derived
Q. Wang, B. Jiang, Y. Ding, X. Zeng, Y.S. Zeng, Tissue-engineered neural network cells induces functional recovery in rats with complete spinal cord transection,
graft relays excitatory signal in the completely transected canine spinal cord, Adv Front. Neurosci. 11 (2017) 589.
Sci (Weinh.Baden-Wurttemberg, Germany) 6 (22) (2019) 1901240. [59] T. Ben-Hur, Reconstructing neural circuits using transplanted neural stem cells in
[36] K.M. Fisher, B. Zaaimi, S.A. Edgley, S.N. Baker, Extensive cortical convergence to the injured spinal cord, J. Clin. Investig. 120 (9) (2010) 3096–3098.
primate reticulospinal pathways, J. Neurosci. : off. J. Soc. Neurosci. 41 (5) (2021) [60] G. Li, M.T. Che, X. Zeng, X.C. Qiu, B. Feng, B.Q. Lai, H.Y. Shen, E.A. Ling, Y.
1005–1018. S. Zeng, Neurotrophin-3 released from implant of tissue-engineered fibroin
[37] B.Q. Lai, M.T. Che, B.L. Du, X. Zeng, Y.H. Ma, B. Feng, X.C. Qiu, K. Zhang, S. Liu, scaffolds inhibits inflammation, enhances nerve fiber regeneration, and improves
H.Y. Shen, J.L. Wu, E.A. Ling, Y.S. Zeng, Transplantation of tissue engineering motor function in canine spinal cord injury, J. Biomed. Mater. Res. 106 (8) (2018)
neural network and formation of neuronal relay into the transected rat spinal 2158–2170.
cord, Biomaterials 109 (2016) 40–54. [61] N.D. James, S.B. McMahon, E.C. Field-Fote, E.J. Bradbury, Neuromodulation in
[38] J.T. Hachmann, P.J. Grahn, J.S. Calvert, D.I. Drubach, K.H. Lee, I.A. Lavrov, the restoration of function after spinal cord injury, Lancet Neurol. 17 (10) (2018)
Electrical neuromodulation of the respiratory system After spinal cord injury, 905–917.
Mayo Clin. Proc. 92 (9) (2017) 1401–1414. [62] Z.C. Gao, B.B. Niu, M.C. Gu, Y.H. Li, J.T. Liu, Y.B. Wang, X.J. He, [Clinical effects
[39] X.Y. Lin, B.Q. Lai, X. Zeng, M.T. Che, E.A. Ling, W. Wu, Y.S. Zeng, Cell of high frequency repeated transcranial magnetic stimulation therapy on
transplantation and neuroengineering approach for spinal cord injury treatment: dyskinesia in patients with incomplete spinal cord injury:a Meta-analysis],
a summary of current laboratory findings and review of literature, Cell Zhongguo gu shang = China j. orthop. traumatol. 31 (1) (2018) 47–55.
Transplant. 25 (8) (2016) 1425–1438. [63] R.D. Wilson, A.M. Bryden, K.L. Kilgore, N. Makowski, D. Bourbeau, K.E. Kowalski,
[40] V.R. Edgerton, G. Courtine, Y.P. Gerasimenko, I. Lavrov, R.M. Ichiyama, A. A.F. DiMarco, J.S. Knutson, Neuromodulation for functional electrical
J. Fong, L.L. Cai, C.K. Otoshi, N.J. Tillakaratne, J.W. Burdick, R.R. Roy, Training stimulation, Phys. Med. Rehabil. Clin 30 (2) (2019) 301–318.
locomotor networks, Brain Res. Rev. 57 (1) (2008) 241–254. [64] K. Yokota, K. Kobayakawa, K. Kubota, A. Miyawaki, H. Okano, Y. Ohkawa,
Y. Iwamoto, S. Okada, Engrafted neural stem/progenitor cells promote functional

14
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

recovery through synapse reorganization with spared host neurons after spinal [89] L.E. Sperling, K. Pires Reis, F. Nicola, C. Euzebio Teixeira, G. Gulielmin Dido,
cord injury, Stem cell reports 5 (2) (2015) 264–277. M. Garrido Dos Santos, E. Konrath, C.A. Netto, P. Pranke, Galantamine improves
[65] M. Cusimano, E. Brambilla, A. Capotondo, D. De Feo, A. Tomasso, G. Comi, functional recovery and reduces lesion size in a rat model of spinal cord injury,
P. D’Adamo, L. Muzio, G. Martino, Selective killing of spinal cord neural stem Brain Res. 1724 (2019) 146424.
cells impairs locomotor recovery in a mouse model of spinal cord injury, [90] M. Stenudd, H. Sabelstrom, J. Frisen, Role of endogenous neural stem cells in
J. Neuroinflammation 15 (1) (2018) 58. spinal cord injury and repair, JAMA Neurol. 72 (2) (2015) 235–237.
[66] J. Shang, H. Qiao, P. Hao, Y. Gao, W. Zhao, H. Duan, Z. Yang, X. Li, bFGF-Sodium [91] C.G. Becker, T. Becker, J.P. Hugnot, The spinal ependymal zone as a source of
hyaluronate collagen scaffolds enable the formation of nascent neural networks endogenous repair cells across vertebrates, Prog. Neurobiol. 170 (2018) 67–80.
after adult spinal cord injury, J. Biomed. Nanotechnol. 15 (4) (2019) 703–716. [92] Q.L. Cao, Y.P. Zhang, R.M. Howard, W.M. Walters, P. Tsoulfas, S.R. Whittemore,
[67] Y.T. Zhang, H. Jin, J.H. Wang, L.Y. Wen, Y. Yang, J.W. Ruan, S.X. Zhang, E. Pluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord
A. Ling, Y. Ding, Y.S. Zeng, Tail nerve electrical stimulation and electro- are restricted to a glial lineage, Exp. Neurol. 167 (1) (2001) 48–58.
acupuncture can protect spinal motor neurons and alleviate muscle atrophy after [93] Z. Ma, Y.P. Zhang, W. Liu, G. Yan, Y. Li, L.B.E. Shields, M. Walker, K. Chen,
spinal cord transection in rats, Neural Plast. 2017 (2017) 7351238. W. Huang, M. Kong, Y. Lu, B. Brommer, X. Chen, X.M. Xu, C.B. Shields,
[68] A.M. Wiersma, K. Fouad, I.R. Winship, Enhancing spinal plasticity amplifies the A controlled spinal cord contusion for the rhesus macaque monkey, Exp. Neurol.
benefits of rehabilitative training and improves recovery from stroke, J. Neurosci. 279 (2016) 261–273.
: off. J. Soc. Neurosci. 37 (45) (2017) 10983–10997. [94] T.M. O’Shea, J.E. Burda, M.V. Sofroniew, Cell biology of spinal cord injury and
[69] H. Duan, W. Song, W. Zhao, Y. Gao, Z. Yang, X. Li, Endogenous neurogenesis in repair, J. Clin. Investig. 127 (9) (2017) 3259–3270.
adult mammals after spinal cord injury, Science China, Life Sci. 59 (12) (2016) [95] Y. Xie, W. Song, W. Zhao, Y. Gao, J. Shang, P. Hao, Z. Yang, H. Duan, X. Li,
1313–1318. Application of the sodium hyaluronate-CNTF scaffolds in repairing adult rat
[70] W. Yin, X. Li, Y. Zhao, J. Tan, S. Wu, Y. Cao, J. Li, H. Zhu, W. Liu, G. Tang, spinal cord injury and facilitating neural network formation, Science China, Life
L. Meng, L. Wang, B. Zhu, G. Wang, M. Zhong, X. Liu, D. Xie, B. Chen, C. Ren, Sci. 61 (5) (2018) 559–568.
Z. Xiao, X. Jiang, J. Dai, Taxol-modified collagen scaffold implantation promotes [96] W. Xue, C. Fan, B. Chen, Y. Zhao, Z. Xiao, J. Dai, Direct neuronal differentiation of
functional recovery after long-distance spinal cord complete transection in neural stem cells for spinal cord injury repair, Stem Cell. 39 (8) (2021)
canines, Biomater. Sci. 6 (5) (2018) 1099–1108. 1025–1032.
[71] X. Li, Z. Xiao, J. Han, L. Chen, H. Xiao, F. Ma, X. Hou, X. Li, J. Sun, W. Ding, [97] T. Yuan, Y. Shao, X. Zhou, Q. Liu, Z. Zhu, B. Zhou, Y. Dong, N. Stephanopoulos,
Y. Zhao, B. Chen, J. Dai, Promotion of neuronal differentiation of neural S. Gui, H. Yan, D. Liu, Highly permeable DNA supramolecular hydrogel promotes
progenitor cells by using EGFR antibody functionalized collagen scaffolds for neurogenesis and functional recovery after completely transected spinal cord
spinal cord injury repair, Biomaterials 34 (21) (2013) 5107–5116. injury, advanced materials (deerfield beach, fla 33 (35) (2021), e2102428.
[72] X. Li, J. Han, Y. Zhao, W. Ding, J. Wei, J. Li, S. Han, X. Shang, B. Wang, B. Chen, [98] M.C. Mosley, H.J. Lim, J. Chen, Y.H. Yang, S. Li, Y. Liu, L.A. Smith Callahan,
Z. Xiao, J. Dai, Functionalized collagen scaffold implantation and cAMP Neurite extension and neuronal differentiation of human induced pluripotent
administration collectively facilitate spinal cord regeneration, Acta Biomater. 30 stem cell derived neural stem cells on polyethylene glycol hydrogels containing a
(2016) 233–245. continuous Young’s Modulus gradient, J. Biomed. Mater. Res. 105 (3) (2017)
[73] X. Li, Y. Zhao, S. Cheng, S. Han, M. Shu, B. Chen, X. Chen, F. Tang, N. Wang, 824–833.
Y. Tu, B. Wang, Z. Xiao, S. Zhang, J. Dai, Cetuximab modified collagen scaffold [99] J.H. Sun, G. Li, T.T. Wu, Z.J. Lin, J.L. Zou, L.J. Huang, H.Y. Xu, J.H. Wang, Y.
directs neurogenesis of injury-activated endogenous neural stem cells for acute H. Ma, Y.S. Zeng, Decellularization optimizes the inhibitory microenvironment of
spinal cord injury repair, Biomaterials 137 (2017) 73–86. the optic nerve to support neurite growth, Biomaterials 258 (2020) 120289.
[74] X. Li, C. Fan, Z. Xiao, Y. Zhao, H. Zhang, J. Sun, Y. Zhuang, X. Wu, J. Shi, Y. Chen, [100] Y.R. Bai, B.Q. Lai, W.T. Han, J.H. Sun, G. Li, Y. Ding, X. Zeng, Y.H. Ma, Y.S. Zeng,
J. Dai, A collagen microchannel scaffold carrying paclitaxel-liposomes induces Decellularized optic nerve functional scaffold transplant facilitates directional
neuronal differentiation of neural stem cells through Wnt/beta-catenin signaling axon regeneration and remyelination in the injured white matter of the rat spinal
for spinal cord injury repair, Biomaterials 183 (2018) 114–127. cord, Neural regen. res. 16 (11) (2021) 2276–2283.
[75] S. Han, B. Wang, W. Jin, Z. Xiao, X. Li, W. Ding, M. Kapur, B. Chen, B. Yuan, [101] P. Lu, G. Woodruff, Y. Wang, L. Graham, M. Hunt, D. Wu, E. Boehle, R. Ahmad,
T. Zhu, H. Wang, J. Wang, Q. Dong, W. Liang, J. Dai, The linear-ordered collagen G. Poplawski, J. Brock, L.S. Goldstein, M.H. Tuszynski, Long-distance axonal
scaffold-BDNF complex significantly promotes functional recovery after growth from human induced pluripotent stem cells after spinal cord injury,
completely transected spinal cord injury in canine, Biomaterials 41 (2015) 89–96. Neuron 83 (4) (2014) 789–796.
[76] L.D. Hachem, A.J. Mothe, C.H. Tator, Unlocking the paradoxical endogenous [102] F.S. Wong, B.P. Chan, A.C. Lo, Carriers in cell-based therapies for neurological
stem cell response after spinal cord injury, Stem Cell. 38 (2) (2020) 187–194. disorders, Int. J. Mol. Sci. 15 (6) (2014) 10669–10723.
[77] H. Li, G. Chen, In vivo reprogramming for CNS repair: regenerating neurons from [103] O. Steward, K.G. Sharp, K. Matsudaira Yee, Long-distance migration and
endogenous glial cells, Neuron 91 (4) (2016) 728–738. colonization of transplanted neural stem cells, Cell 156 (3) (2014) 385–387.
[78] L.L. Wang, Z. Su, W. Tai, Y. Zou, X.M. Xu, C.L. Zhang, The p53 pathway controls [104] H. Kanno, D.D. Pearse, H. Ozawa, E. Itoi, M.B. Bunge, Schwann cell
SOX2-mediated reprogramming in the adult mouse spinal cord, Cell Rep. 17 (3) transplantation for spinal cord injury repair: its significant therapeutic potential
(2016) 891–903. and prospectus, Rev. Neurosci. 26 (2) (2015) 121–128.
[79] J. Mertens, M.C. Marchetto, C. Bardy, F.H. Gage, Evaluating cell reprogramming, [105] O. Steward, K.G. Sharp, K.M. Yee, M.N. Hatch, J.F. Bonner, Characterization of
differentiation and conversion technologies in neuroscience, Nat. Rev. Neurosci. ectopic colonies that form in widespread areas of the nervous system with neural
17 (7) (2016) 424–437. stem cell transplants into the site of a severe spinal cord injury, J. Neurosci. : off.
[80] T. Matsuda, T. Irie, S. Katsurabayashi, Y. Hayashi, T. Nagai, N. Hamazaki, A.M. J. Soc. Neurosci. 34 (42) (2014) 14013–14021.
D. Adefuin, F. Miura, T. Ito, H. Kimura, K. Shirahige, T. Takeda, K. Iwasaki, [106] Y. Li, W.M. Zhang, T.H. Wang, Optimal location and time for neural stem cell
T. Imamura, K. Nakashima, Pioneer factor NeuroD1 rearranges transcriptional transplantation into transected rat spinal cord, Cell. Mol. Neurobiol. 31 (3) (2011)
and epigenetic profiles to execute microglia-neuron conversion, Neuron 101 (3) 407–414.
(2019) 472–485 e7. [107] J.M. Wang, Y.S. Zeng, J.L. Wu, Y. Li, Y.D. Teng, Cograft of neural stem cells and
[81] K. Jin, M. Zou, D. Xiao, M. Xiang, Reprogramming fibroblasts to neural stem cells schwann cells overexpressing TrkC and neurotrophin-3 respectively after rat
by overexpression of the transcription factor Ptf1a, methods in molecular biology spinal cord transection, Biomaterials 32 (30) (2011) 7454–7468.
(clifton, N. J. 2117 (2020) 245–263. [108] J. Zhang, X. Lu, G. Feng, Z. Gu, Y. Sun, G. Bao, G. Xu, Y. Lu, J. Chen, L. Xu,
[82] G. Chen, M. Wernig, B. Berninger, M. Nakafuku, M. Parmar, C.L. Zhang, In vivo X. Feng, Z. Cui, Chitosan scaffolds induce human dental pulp stem cells to neural
reprogramming for brain and spinal cord repair, eNeuro 2 (5) (2015). differentiation: potential roles for spinal cord injury therapy, Cell Tissue Res. 366
[83] R.A. Barker, M. Gotz, M. Parmar, New approaches for brain repair-from rescue to (1) (2016) 129–142.
reprogramming, Nature 557 (7705) (2018) 329–334. [109] B.Q. Lai, J.M. Wang, E.A. Ling, J.L. Wu, Y.S. Zeng, Graft of a tissue-engineered
[84] Y. Guo, S. Liu, X. Zhang, L. Wang, X. Zhang, A. Hao, A. Han, J. Yang, Sox11 neural scaffold serves as a promising strategy to restore myelination after rat
promotes endogenous neurogenesis and locomotor recovery in mice spinal cord spinal cord transection, Stem Cell. Dev. 23 (8) (2014) 910–921.
injury, Biochem. Biophys. Res. Commun. 446 (4) (2014) 830–835. [110] Y. Xiong, Y.S. Zeng, C.G. Zeng, B.L. Du, L.M. He, D.P. Quan, W. Zhang, J.
[85] L. Yang, G. Li, J. Ye, D. Lu, Z. Chen, A.P. Xiang, M.H. Jiang, Substance P enhances M. Wang, J.L. Wu, Y. Li, J. Li, Synaptic transmission of neural stem cells seeded in
endogenous neurogenesis to improve functional recovery after spinal cord injury, 3-dimensional PLGA scaffolds, Biomaterials 30 (22) (2009) 3711–3722.
Int. J. Biochem. Cell Biol. 89 (2017) 110–119. [111] Y. Lee, B.M. Morrison, Y. Li, S. Lengacher, M.H. Farah, P.N. Hoffman, Y. Liu,
[86] C. Fan, X. Li, Z. Xiao, Y. Zhao, H. Liang, B. Wang, S. Han, X. Li, B. Xu, N. Wang, A. Tsingalia, L. Jin, P.W. Zhang, L. Pellerin, P.J. Magistretti, J.D. Rothstein,
S. Liu, W. Xue, J. Dai, A modified collagen scaffold facilitates endogenous Oligodendroglia metabolically support axons and contribute to
neurogenesis for acute spinal cord injury repair, Acta Biomater. 51 (2017) neurodegeneration, Nature 487 (7408) (2012) 443–448.
304–316. [112] Y. Yang, H.Y. Xu, Q.W. Deng, G.H. Wu, X. Zeng, H. Jin, L.J. Wang, B.Q. Lai, G. Li,
[87] G. Li, M.T. Che, K. Zhang, L.N. Qin, Y.T. Zhang, R.Q. Chen, L.M. Rong, S. Liu, Y.H. Ma, B. Jiang, J.W. Ruan, Y.Q. Wang, Y. Ding, Y.S. Zeng, Electroacupuncture
Y. Ding, H.Y. Shen, S.M. Long, J.L. Wu, E.A. Ling, Y.S. Zeng, Graft of the NT-3 facilitates the integration of a grafted TrkC-modified mesenchymal stem cell-
persistent delivery gelatin sponge scaffold promotes axon regeneration, derived neural network into transected spinal cord in rats via increasing
attenuates inflammation, and induces cell migration in rat and canine with spinal neurotrophin-3, CNS Neurosci. Ther. 27 (7) (2021) 776–791.
cord injury, Biomaterials 83 (2016) 233–248. [113] A. Marchini, A. Raspa, R. Pugliese, M.A. El Malek, V. Pastori, M. Lecchi, A.
[88] L. Zhou, L. Fan, X. Yi, Z. Zhou, C. Liu, R. Fu, C. Dai, Z. Wang, X. Chen, P. Yu, L. Vescovi, F. Gelain, Multifunctionalized hydrogels foster hNSC maturation in 3D
D. Chen, G. Tan, Q. Wang, C. Ning, Soft conducting polymer hydrogels cross- cultures and neural regeneration in spinal cord injuries, Proc. Natl. Acad. Sci. U S
linked and doped by tannic acid for spinal cord injury repair, ACS Nano 12 (11) A. 116 (15) (2019) 7483–7492.
(2018) 10957–10967. [114] J. Wang, W. Zou, J. Ma, J. Liu, Biomaterials and gene manipulation in stem cell-
based therapies for spinal cord injury, Stem Cell. Dev. 28 (4) (2019) 239–257.

15
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

[115] J.M. Wang, Y.S. Zeng, R.Y. Liu, W.L. Huang, Y. Xiong, Y.H. Wang, S.J. Chen, Y. [135] A. Roshani, A. Erfanian, A modular robust control framework for control of
D. Teng, Recombinant adenovirus vector-mediated functional expression of movement elicited by multi-electrode intraspinal microstimulation, J. Neural.
neurotropin-3 receptor (TrkC) in neural stem cells, Exp. Neurol. 203 (1) (2007) Eng. 13 (4) (2016), 046024.
123–127. [136] A. Roshani, A. Erfanian, The effects of stimulation strategy on joint movement
[116] X. Zeng, Y.S. Zeng, Y.H. Ma, L.Y. Lu, B.L. Du, W. Zhang, Y. Li, W.Y. Chan, Bone elicited by intraspinal microstimulation, IEEE Trans. Neural Syst. Rehabil. Eng. :
marrow mesenchymal stem cells in a three-dimensional gelatin sponge scaffold Conf Proc IEEE Eng Med Biol Soc 24 (7) (2016) 794–805.
attenuate inflammation, promote angiogenesis, and reduce cavity formation in [137] A. Roshani, A. Erfanian, Fuzzy logic control of ankle movement using multi-
experimental spinal cord injury, Cell Transplant. 20 (11–12) (2011) 1881–1899. electrode intraspinal microstimulation, Conference proceedings, in: Annual
[117] G. Li, Y. Cao, F. Shen, Y. Wang, L. Bai, W. Guo, Y. Bi, G. Lv, Z. Fan, Mdivi-1 International Conference of the IEEE Engineering in Medicine and Biology
inhibits astrocyte activation and astroglial scar formation and enhances axonal Society. IEEE Engineering in Medicine and Biology Society. Annual Conference
regeneration after spinal cord injury in rats, Front. Cell. Neurosci. 10 (2016) 241. 2013, 2013, pp. 5642–5645.
[118] J.L. Zou, S. Liu, J.H. Sun, W.H. Yang, Y.W. Xu, Z.L. Rao, B. Jiang, Q.T. Zhu, X. [138] J.B. Zimmermann, K. Seki, A. Jackson, Reanimating the arm and hand with
L. Liu, J.L. Wu, C. Chang, H.Q. Mao, E.A. Ling, D.P. Quan, Y.S. Zeng, Peripheral intraspinal microstimulation, J. Neural. Eng. 8 (5) (2011), 054001.
nerve-derived matrix hydrogel promotes remyelination and inhibits synapse [139] L.M. Mercier, E.J. Gonzalez-Rothi, K.A. Streeter, S.S. Posgai, A.S. Poirier, D.
formation, Adv. Funct. Mater. 28 (13) (2018). D. Fuller, P.J. Reier, D.M. Baekey, Intraspinal microstimulation and diaphragm
[119] S. Boncompagni, H. Kern, K. Rossini, C. Hofer, W. Mayr, U. Carraro, F. Protasi, activation after cervical spinal cord injury, J. Neurophysiol. 117 (2) (2017)
Structural differentiation of skeletal muscle fibers in the absence of innervation in 767–776.
humans, Proc. Natl. Acad. Sci. U S A. 104 (49) (2007) 19339–19344. [140] A.N. Dalrymple, D.G. Everaert, D.S. Hu, V.K. Mushahwar, A speed-adaptive
[120] R.E. Thompson, J. Pardieck, L. Smith, P. Kenny, L. Crawford, M. Shoichet, intraspinal microstimulation controller to restore weight-bearing stepping in a
S. Sakiyama-Elbert, Effect of hyaluronic acid hydrogels containing astrocyte- spinal cord hemisection model, J. Neural. Eng. 15 (5) (2018), 056023.
derived extracellular matrix and/or V2a interneurons on histologic outcomes [141] V.K. Mushahwar, L. Guevremont, R. Saigal, Could cortical signals control
following spinal cord injury, Biomaterials 162 (2018) 208–223. intraspinal stimulators? A theoretical evaluation, IEEE Trans. Neural Syst.
[121] N. Wenger, E.M. Moraud, J. Gandar, P. Musienko, M. Capogrosso, L. Baud, C.G. Le Rehabil. Eng. : Conf Proc IEEE Eng Med Biol Soc 14 (2) (2006) 198–201.
Goff, Q. Barraud, N. Pavlova, N. Dominici, I.R. Minev, L. Asboth, A. Hirsch, [142] F. Panetsos, A. Sanchez-Jimenez, C. Torets, C. Largo, S. Micera, Controlling
S. Duis, J. Kreider, A. Mortera, O. Haverbeck, S. Kraus, F. Schmitz, J. DiGiovanna, selective stimulations below a spinal cord hemisection using brain recordings
R. van den Brand, J. Bloch, P. Detemple, S.P. Lacour, E. Bezard, S. Micera, with a neural interface system approach, J. Neural. Eng. 8 (4) (2011), 046008.
G. Courtine, Spatiotemporal neuromodulation therapies engaging muscle [143] E. Rejc, C.A. Angeli, Spinal cord epidural stimulation for lower limb motor
synergies improve motor control after spinal cord injury, Nat. Med. 22 (2) (2016) function recovery in individuals with motor complete spinal cord injury, Phys.
138–145. Med. Rehabil. Clin 30 (2) (2019) 337–354.
[122] M. Capogrosso, T. Milekovic, D. Borton, F. Wagner, E.M. Moraud, J.B. Mignardot, [144] J.G. Xu, Y.Q. Tu, Y.D. Gu, [Effect of electric stimulation on denervated skeletal
N. Buse, J. Gandar, Q. Barraud, D. Xing, E. Rey, S. Duis, Y. Jianzhong, W.K. Ko, muscle atrophy], Zhongguo xiu fu chong jian wai ke za zhi = Zhongguo xiufu
Q. Li, P. Detemple, T. Denison, S. Micera, E. Bezard, J. Bloch, G. Courtine, chongjian waike zazhi = Chin. j. reparative reconstr. surg. 17 (5) (2003)
A brain-spine interface alleviating gait deficits after spinal cord injury in 396–399.
primates, Nature 539 (7628) (2016) 284–288. [145] H. Kern, S. Boncompagni, K. Rossini, W. Mayr, G. Fano, M.E. Zanin,
[123] M. Bonizzato, G. Pidpruzhnykova, J. DiGiovanna, P. Shkorbatova, N. Pavlova, M. Podhorska-Okolow, F. Protasi, U. Carraro, Long-term denervation in humans
S. Micera, G. Courtine, Brain-controlled modulation of spinal circuits improves causes degeneration of both contractile and excitation-contraction coupling
recovery from spinal cord injury, Nat. Commun. 9 (1) (2018) 3015. apparatus, which is reversible by functional electrical stimulation (FES): a role for
[124] R. van den Brand, J. Heutschi, Q. Barraud, J. DiGiovanna, K. Bartholdi, myofiber regeneration? J. Neuropathol. Exp. Neurol. 63 (9) (2004) 919–931.
M. Huerlimann, L. Friedli, I. Vollenweider, E.M. Moraud, S. Duis, N. Dominici, [146] U. Carraro, S. Boncompagni, V. Gobbo, K. Rossini, S. Zampieri, S. Mosole,
S. Micera, P. Musienko, G. Courtine, Restoring voluntary control of locomotion B. Ravara, A. Nori, R. Stramare, F. Ambrosio, F. Piccione, S. Masiero, V. Vindigni,
after paralyzing spinal cord injury, Science (New York, N.Y.) 336 (6085) (2012) P. Gargiulo, F. Protasi, H. Kern, A. Pond, A. Marcante, Persistent muscle fiber
1182–1185. regeneration in long term denervation, Past, Present, Future. Eur. J. Transl. Myol.
[125] F.B. Wagner, J.B. Mignardot, C.G. Le Goff-Mignardot, R. Demesmaeker, S. Komi, 25 (2) (2015) 4832.
M. Capogrosso, A. Rowald, I. Seanez, M. Caban, E. Pirondini, M. Vat, L. [147] N. Hayashi, N. Himi, E. Nakamura-Maruyama, N. Okabe, I. Sakamoto,
A. McCracken, R. Heimgartner, I. Fodor, A. Watrin, P. Seguin, E. Paoles, K. Van T. Hasegawa, O. Miyamoto, Improvement of motor function induced by skeletal
Den Keybus, G. Eberle, B. Schurch, E. Pralong, F. Becce, J. Prior, N. Buse, muscle contraction in spinal cord-injured rats, Spine J. : off. J. North Am. Spine
R. Buschman, E. Neufeld, N. Kuster, S. Carda, J. von Zitzewitz, V. Delattre, Soc. 19 (6) (2019) 1094–1105.
T. Denison, H. Lambert, K. Minassian, J. Bloch, G. Courtine, Targeted [148] K. Weber, Neuromodulation and devices in trigeminal neuralgia, Headache 57
neurotechnology restores walking in humans with spinal cord injury, Nature 563 (10) (2017) 1648–1653.
(7729) (2018) 65–71. [149] S. Wurth, M. Capogrosso, S. Raspopovic, J. Gandar, G. Federici, N. Kinany,
[126] J. Ilha, A. Meireles, G.R. de Freitas, C.C. do Espirito Santo, N. Machado-Pereira, A. Cutrone, A. Piersigilli, N. Pavlova, R. Guiet, G. Taverni, J. Rigosa,
A. Swarowsky, A.R.S. Santos, Overground gait training promotes functional P. Shkorbatova, X. Navarro, Q. Barraud, G. Courtine, S. Micera, Long-term
recovery and cortical neuroplasticity in an incomplete spinal cord injury model, usability and bio-integration of polyimide-based intra-neural stimulating
Life Sci. 232 (2019) 116627. electrodes, Biomaterials 122 (2017) 114–129.
[127] S. Harkema, Y. Gerasimenko, J. Hodes, J. Burdick, C. Angeli, Y. Chen, C. Ferreira, [150] E. Formento, K. Minassian, F. Wagner, J.B. Mignardot, C.G. Le Goff-Mignardot,
A. Willhite, E. Rejc, R.G. Grossman, V.R. Edgerton, Effect of epidural stimulation A. Rowald, J. Bloch, S. Micera, M. Capogrosso, G. Courtine, Electrical spinal cord
of the lumbosacral spinal cord on voluntary movement, standing, and assisted stimulation must preserve proprioception to enable locomotion in humans with
stepping after motor complete paraplegia: a case study, Lancet (London, England) spinal cord injury, Nat. Neurosci. 21 (12) (2018) 1728–1741.
377 (9781) (2011) 1938–1947. [151] K. Zheng, G. Feng, J. Zhang, J. Xing, D. Huang, M. Lian, W. Zhang, W. Wu, Y. Hu,
[128] S. Sharififar, J.J. Shuster, M.D. Bishop, Adding electrical stimulation during X. Lu, X. Feng, Basic fibroblast growth factor promotes human dental pulp stem
standard rehabilitation after stroke to improve motor function. A systematic cells cultured in 3D porous chitosan scaffolds to neural differentiation, Int. J.
review and meta-analysis, Ann. Phys. Rehabil. Med. 61 (5) (2018) 339–344. Neurosci. (2020) 1–9.
[129] M.R. Kasten, M.D. Sunshine, E.S. Secrist, P.J. Horner, C.T. Moritz, Therapeutic [152] I. Ammendrup-Johnsen, Y. Naito, A.M. Craig, H. Takahashi, Neurotrophin-3
intraspinal microstimulation improves forelimb function after cervical contusion enhances the synaptic organizing function of TrkC-protein tyrosine phosphatase
injury, J. Neural. Eng. 10 (4) (2013), 044001. sigma in rat hippocampal neurons, J. Neurosci. : off. J. Soc. Neurosci. 35 (36)
[130] B.J. Holinski, K.A. Mazurek, D.G. Everaert, A. Toossi, A.M. Lucas-Osma, P. Troyk, (2015) 12425–12431.
R. Etienne-Cummings, R.B. Stein, V.K. Mushahwar, Intraspinal microstimulation [153] K.A. Han, D. Woo, S. Kim, G. Choii, S. Jeon, S.Y. Won, H.M. Kim, W.D. Heo, J.
produces over-ground walking in anesthetized cats, J. Neural. Eng. 13 (5) (2016), W. Um, J. Ko, Neurotrophin-3 regulates synapse development by modulating
056016. TrkC-PTPsigma synaptic adhesion and intracellular signaling pathways,
[131] C. Tao, X. Shen, L. Ma, J. Shen, Z. Li, Z. Wang, X. Lu, Comparative study of J. Neurosci. : off. J. Soc. Neurosci. 36 (17) (2016) 4816–4831.
intraspinal microstimulation and epidural spinal cord stimulation, conference [154] Y. Ding, Q. Yan, J.W. Ruan, Y.Q. Zhang, W.J. Li, X. Zeng, S.F. Huang, Y.J. Zhang,
proceedings, in: Annual International Conference of the IEEE Engineering in J.L. Wu, D. Fisher, H. Dong, Y.S. Zeng, Electroacupuncture promotes the
Medicine and Biology Society. IEEE Engineering in Medicine and Biology Society. differentiation of transplanted bone marrow mesenchymal stem cells
Annual Conference 2019, 2019, pp. 3795–3798. overexpressing TrkC into neuron-like cells in transected spinal cord of rats, Cell
[132] J.A. Bamford, V.K. Mushahwar, Intraspinal microstimulation for the recovery of Transplant. 22 (1) (2013) 65–86.
function following spinal cord injury, Prog. Brain Res. 194 (2011) 227–239. [155] A.B. Ajiboye, F.R. Willett, D.R. Young, W.D. Memberg, B.A. Murphy, J.P. Miller,
[133] P.J. Grahn, G.W. Mallory, B.M. Berry, J.T. Hachmann, D.A. Lobel, J.L. Lujan, B.L. Walter, J.A. Sweet, H.A. Hoyen, M.W. Keith, P.H. Peckham, J.D. Simeral, J.
Restoration of motor function following spinal cord injury via optimal control of P. Donoghue, L.R. Hochberg, R.F. Kirsch, Restoration of reaching and grasping
intraspinal microstimulation: toward a next generation closed-loop neural movements through brain-controlled muscle stimulation in a person with
prosthesis, Front. Neurosci. 8 (2014) 296. tetraplegia: a proof-of-concept demonstration, Lancet (London, England) 389
[134] S. Shahdoost, S. Frost, C. Dunham, S. DeJong, S. Barbay, R. Nudo, P. Mohseni, (10081) (2017) 1821–1830.
Cortical control of intraspinal microstimulation: toward a new approach for [156] C.A. Angeli, M. Boakye, R.A. Morton, J. Vogt, K. Benton, Y. Chen, C.K. Ferreira, S.
restoration of function after spinal cord injury, Conference proceedings, in: J. Harkema, Recovery of over-ground walking after chronic motor complete
Annual International Conference of the IEEE Engineering in Medicine and Biology spinal cord injury, N. Engl. J. Med. 379 (13) (2018) 1244–1250.
Society. IEEE Engineering in Medicine and Biology Society. Annual Conference [157] U.S. Hofstoetter, M. Krenn, S.M. Danner, C. Hofer, H. Kern, W.B. McKay, W. Mayr,
2015, 2015, pp. 2159–2162. K. Minassian, Augmentation of voluntary locomotor activity by transcutaneous

16
B.-Q. Lai et al. Biomaterials 279 (2021) 121211

spinal cord stimulation in motor-incomplete spinal cord-injured individuals, Artif. [168] S.J. Liu, S.S. Zheng, Q.Q. Dan, J. Liu, T.H. Wang, Effects of Governor Vessel
Organs 39 (10) (2015) E176–E186. electroacupuncture on the systematic expressions of NTFs in spinal cord
[158] S.P. Estes, J.A. Iddings, E.C. Field-Fote, Priming neural circuits to modulate spinal transected rats, Neuropeptides 48 (4) (2014) 239–247.
reflex excitability, Front. Neurol. 8 (2017) 17. [169] S. Canavero, X.P. Ren, The spark of life: engaging the cortico-truncoreticulo-
[159] D. Teoli, J. an, Transcutaneous Electrical Nerve Stimulation (TENS), StatPearls, propriospinal pathway by electrical stimulation, CNS Neurosci. Ther. 22 (4)
StatPearls Publishing LLC., Treasure Island FL, 2018. (2016) 260–261.
[160] Y.X. Zhou, H.P. Wang, X.L. Bao, X.Y. Lu, Z.G. Wang, A frequency and pulse-width [170] X. Wang, S. Ju, S. Chen, W. Gao, J. Ding, G. Wang, H. Cao, H. Tian, X. Li, Effect of
co-modulation strategy for transcutaneous neuromuscular electrical stimulation electro-acupuncture on neuroplasticity of spinal cord-transected rats, Med. Sci.
based on sEMG time-domain features, J. Neural. Eng. 13 (1) (2016), 016004. Mon. Int. Med. J. Exp. Clin. Res. : Int. Med. J. Exp. Clin. Res 23 (2017)
[161] W. Mayr, M. Krenn, M.R. Dimitrijevic, Epidural and transcutaneous spinal 4241–4251.
electrical stimulation for restoration of movement after incomplete and complete [171] T.H. Hutson, C. Kathe, I. Palmisano, K. Bartholdi, A. Hervera, F. De Virgiliis,
spinal cord injury, Curr. Opin. Neurol. 29 (6) (2016) 721–726. E. McLachlan, L. Zhou, G. Kong, Q. Barraud, M.C. Danzi, A. Medrano-Fernandez,
[162] A. Chari, I.D. Hentall, M.C. Papadopoulos, E.A. Pereira, Surgical neurostimulation J.P. Lopez-Atalaya, A.L. Boutillier, S.H. Sinha, A.K. Singh, P. Chaturbedy, L.D.
for spinal cord injury, Brain Sci. 7 (2) (2017). F. Moon, T.K. Kundu, J.L. Bixby, V.P. Lemmon, A. Barco, G. Courtine, S. Di
[163] A.A. Mahrous, M.H. Mousa, S.M. Elbasiouny, The mechanistic basis for successful Giovanni, Cbp-dependent histone acetylation mediates axon regeneration
spinal cord stimulation to generate steady motor outputs, Front. Cell. Neurosci. induced by environmental enrichment in rodent spinal cord injury models, Sci.
13 (2019) 359. Transl. Med. 11 (487) (2019).
[164] E.S. Hong, H.H. Yao, Y.J. Min, J. Sun, X. Zhou, X.B. Zeng, W. Yu, The mechanism [172] M. Capogrosso, N. Wenger, S. Raspopovic, P. Musienko, J. Beauparlant, L. Bassi
of electroacupuncture for treating spinal cord injury rats by mediating Rho/Rho- Luciani, G. Courtine, S. Micera, A computational model for epidural electrical
associated kinase signaling pathway, J Spinal Cord Med (2019) 1–11. stimulation of spinal sensorimotor circuits, J. Neurosci. : off. J. Soc. Neurosci. 33
[165] H. Xu, Y. Yang, Q.W. Deng, B.B. Zhang, J.W. Ruan, H. Jin, J.H. Wang, J. Ren, (49) (2013) 19326–19340.
B. Jiang, J.H. Sun, Y.S. Zeng, Y. Ding, Governor vessel electro-acupuncture [173] M.L. Gill, P.J. Grahn, J.S. Calvert, M.B. Linde, I.A. Lavrov, J.A. Strommen, L.
promotes the intrinsic growth ability of spinal neurons through activating A. Beck, D.G. Sayenko, M.G. Van Straaten, D.I. Drubach, D.D. Veith, A.
calcitonin gene-related peptide/α-calcium/calmodulin-dependent protein kinase/ R. Thoreson, C. Lopez, Y.P. Gerasimenko, V.R. Edgerton, K.H. Lee, K.D. Zhao,
neurotrophin-3 pathway after spinal cord injury, J. Neurotrauma 38 (6) (2021) Neuromodulation of lumbosacral spinal networks enables independent stepping
734–745. after complete paraplegia, Nat. Med. 24 (11) (2018) 1677–1682.
[166] W.P. Xiao, L.L. Ding, Y.J. Min, H.Y. Yang, H.H. Yao, J. Sun, X. Zhou, X.B. Zeng, [174] L.A. Harvey, J.V. Glinsky, J.L. Bowden, The effectiveness of 22 commonly
W. Yu, Electroacupuncture promoting axonal regeneration in spinal cord injury administered physiotherapy interventions for people with spinal cord injury: a
rats via suppression of nogo/NgR and rho/ROCK signaling pathway, systematic review, Spinal Cord 54 (11) (2016) 914–923.
Neuropsychiatric Dis. Treat. 15 (2019) 3429–3442. [175] H.H. Liu, Y. Xiang, T.B. Yan, Z.M. Tan, S.H. Li, X.K. He, Functional electrical
[167] T.H. Wang, X.Y. Wang, X.L. Li, H.M. Chen, L.F. Wu, Effect of electroacupuncture stimulation increases neural stem/progenitor cell proliferation and neurogenesis
on neurotrophin expression in cat spinal cord after partial dorsal rhizotomy, in the subventricular zone of rats with stroke, Chin. Med. J. 126 (12) (2013)
Neurochem. Res. 32 (8) (2007) 1415–1422. 2361–2367.
[176] W. Young, Electrical stimulation and motor recovery, Cell Transplant. 24 (3)
(2015) 429–446.

17

You might also like