You are on page 1of 12

Experimental Neurology 369 (2023) 114543

Contents lists available at ScienceDirect

Experimental Neurology
journal homepage: www.elsevier.com/locate/yexnr

Review Article

Stem cells in central nervous system diseases: Promising


therapeutic strategies
Caidi Ying a, b, 1, Jiahao Zhang a, b, 1, Haocheng Zhang a, b, 1, Shiqi Gao a, b, Xiaoming Guo a, b,
Jun Lin a, b, Haijian Wu a, b, *, Yuan Hong a, b, *
a
Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
b
Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China

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

Keywords: Central nervous system (CNS) diseases are a leading cause of death and disability. Due to CNS neurons have no
Stem cell self-renewal and regenerative ability as they mature, their loss after injury or disease is irreversible and often
Cell therapy leads to functional impairments. Unfortunately, therapeutic options for CNS diseases are still limited, and
Central nervous system diseases
effective treatments for these notorious diseases are warranted to be explored. At present, stem cell therapy has
Immunoregulation
Neurotrophic effect
emerged as a potential therapeutic strategy for improving the prognosis of CNS diseases. Accumulating pre­
Cell replacement clinical and clinical evidences have demonstrated that multiple molecular mechanisms, such as cell replacement,
immunoregulation and neurotrophic effect, underlie the use of stem cell therapy for CNS diseases. However,
several issues have yet to be addressed to support its clinical application. Thus, this review article aims to
summarize the role and underlying mechanisms of stem cell therapy in treating CNS diseases. And it is worthy of
further evaluation for the potential therapeutic applications of stem cell treatment in CNS disease.

1. Introduction stroke, spinal cord injury (SCI), and traumatic brain injury (TBI) trigger
a cascade of detrimental events that profoundly impact neurological
CNS diseases are among the most prevalent disorders affecting the health(Uyeda and Muramatsu, 2020). Consequently, there is a pressing
nervous system, leading to irreversible damage and loss of functionality need for therapeutic strategies that can facilitate the restoration of
(Menorca et al., 2013; Walshe, 2014). These diseases are associated with neurological function in the CNS.
high mortality and disability rates, imposing a significant burden on Stem cell therapy has garnered considerable attention as a prospec­
society(Feigin et al., 2021). Traditionally, it was believed that the neu­ tive treatment modality for CNS diseases. The utilized stem cells
rons in the CNS of adult mammals lacked the ability to regenerate encompass embryonic stem cells (ESCs), induced pluripotent stem cells
spontaneously following injury(Huebner and Strittmatter, 2009). (iPSCs), neural stem cells (NSCs), mesenchymal stem cells (MSCs), and
Furthermore, the therapeutic options for CNS diseases are often other stem cell variants(Liau et al., 2020; Zhou et al., 2021; Huang et al.,
impeded by the complex and incompletely understood mechanisms 2021). Extensive research has focused on investigating the application
underlying their pathogenesis(Sakowski and Chen, 2022). Neurode­ of stem cell therapy in numerous neurodegenerative diseases and CNS
generative diseases, in particular, involve intricate interactions among injuries, with relatively limited exploration in the context of tumors and
multiple cell types and metabolic processes within a complex microen­ infections(Huo et al., 2014; Gao et al., 2020). Extensive investigations
vironment(Dugger and Dickson, 2017). Similarly, CNS injuries such as have delved into the potential mechanisms underlying the protective

Abbreviations: CNS, central nervous system; SCI, spinal cord injury; TBI, traumatic brain injury; ESCs, embryonic stem cells; iPSCs, induced pluripotent stem cells;
NSCs, neural stem cells; MSCs, mesenchymal stem cells; PD, Parkinson’s disease; ALS, amyotrophic lateral sclerosis; SVZ, subventricular zone; AD, Alzheimer’s
disease; BM-MSCs, bone marrow-derived mesenchymal stem cells; MUSE cells, Multilineage-Differentiating Stress-Enduring Cells; DA, dopamine; Aβ, amyloid-β;
AChEI, acetylcholinesterase inhibitors; UMN, upper motor neurons; LMN, lower motor neurons; MNs, motor neurons; AMSCs, amnion-derived mesenchymal stem
cells; NGF, nerve growth factor; BDNF, brain-derived neurotrophic factor; GDNF, glial cell line-derived neurotrophic factor; NT-3, neurotrophin-3; ROS, reactive
oxygen species; iNPCs, induced neural progenitor cells.
* Corresponding authors at: Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
E-mail addresses: haijwu@zju.edu.cn (H. Wu), hy0904@zju.edu.cn (Y. Hong).
1
Drs. Caidi Ying, Jiahao Zhang and Haocheng Zhang contributed equally.

https://doi.org/10.1016/j.expneurol.2023.114543
Received 9 July 2023; Received in revised form 11 September 2023; Accepted 14 September 2023
Available online 22 September 2023
0014-4886/© 2023 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
C. Ying et al. Experimental Neurology 369 (2023) 114543

role of stem cells, including their secretory function, anti-inflammatory mesenchymal stem cells (BM-MSCs) have been extensively investigated
properties, and capacity for cellular replacement(Xian et al., 2019; De and demonstrated their capacity to differentiate into neurons or glial
Gioia et al., 2020; Ford et al., 2020). cells, facilitating the replacement of damaged cells in brain tissue
This review examines the pathological characteristics of CNS dis­ (Alexanian et al., 2008; Yan et al., 2013). Additionally, MSCs exert a
eases as potential targets for stem cell therapy. The types of stem cells neuroprotective function through the secretion of neurotrophic growth
currently used in stem cell therapy and their neuroprotective mecha­ factors and other mechanisms(Wakabayashi et al., 2010). The mecha­
nisms are also discussed. The review analyzes the limited achievements nisms underlying the therapeutic effects of MSCs have been widely
and future development trends of stem cell therapy, addressing the ex­ explored in neurological disorders, such as TBI and SCI(Wang et al.,
pectations and concerns of clinical application. 2017; Shao et al., 2019).

2. Stem cell therapies for central nervous system diseases 2.1.5. Meninges-derived stem cells
Meninges-derived stem cells, a specialized category of stem cells
2.1. Types of stem cells originating from the meninges, exhibit remarkable capabilities in
generating diverse neural cell types, encompassing neurons and glial
2.1.1. Embryonic stem cells cells(Bifari et al., 2017; Đặng et al., 2019). Their innate regenerative
ESCs, derived from the inner cell mass of developing embryos, have potential implies a pivotal role in restoring and rejuvenating impaired or
the potential to differentiate into various cell types(Martínez-Morales degenerated neural tissues within the central nervous system, holding
et al., 2013). Their transplantation in stem cell therapy is utilized for significant promise for advancing innovative therapeutic approaches
CNS diseases such as Parkinson’s disease (PD) and SCI(Freed et al., addressing a range of neurological disorders, including stroke and SCI.
2001; Shao et al., 2019). The differentiation of ESCs into neural pre­ (Decimo et al., 2011; Nakagomi et al., 2012). These stem cells constitute
cursor somatic cells, neural lineage cells, NSCs, and other secondary an intriguing and relatively unexplored realm of research, representing
cells can be induced through interactions with bone marrow stromal previously overlooked sources of in vivo nerve regeneration, and they
cells, small molecule inhibitors, or other methods(Drury-Stewart et al., may exert a profound influence on the evolution of stem cell-based
2013). Ethical concerns, tumorigenicity, immune rejection, and other therapies for neurological diseases.
related issues surrounding the application of ESCs have been widely
discussed, limiting their use(Ratajczak et al., 2019). 2.1.6. Other types of stem cells
There are some other types of stem cells, such as Multilineage-
2.1.2. Induced pluripotent stem cells Differentiating Stress-Enduring Cells (MUSE cells), Dental Pulp Stem
iPSCs are generated by reprogramming adult cells into an embryoid Cells, Hematopoietic Stem Cells (CD34 Positive), Endothelial Progenitor
state, allowing them to differentiate into various cell types(Shahbazi Cells and Neural Crest Stem Cells (NCSCs), that have also been used to
et al., 2018). iPSCs share similar advantages with ESCs as they possess study the mechanisms of nerve regeneration in models such as stroke
pluripotency and self-renewal capabilities(Takahashi and Yamanaka, and SCI(Hennemann et al., 2008; Sakai et al., 2012; Park et al., 2014;
2006; Phanstiel et al., 2011). In comparison to ESCs, iPSCs offer benefits Müller et al., 2015; Uchida et al., 2016). At present, there are few studies
such as ease of acquisition, ease of cultivation, and reduced immuno­ on these stem cells in CNS diseases, and they will not be introduced in
genicity(Vitrac and Cloëz-Tayarani, 2018). iPSCs find application in detail in this review.
stem cell therapy for CNS diseases like amyotrophic lateral sclerosis
(ALS) and SCI(Kim et al., 2014; Shao et al., 2019). However, it is 2.2. Stem cell therapies for neurodegenerative diseases
important to note that while iPSC differentiation into neurons or glial
cells may enhance the efficacy of stem cell therapy, there is a potential Stem cell therapy has emerged as a valuable approach in the treat­
risk of tumorigenicity(Suda et al., 2020). ment of various CNS diseases, particularly neurodegenerative diseases.
Neurodegenerative diseases are characterized by the progressive
2.1.3. Neural stem cells degeneration and loss of neurons in terms of structure, function, or
NSCs possess the capability to differentiate into neurons, astrocytes, quantity(Yuan and Yankner, 2000). PD, AD, and ALS are the three main
and oligodendrocytes(Urbán et al., 2019). In adult mammals, NSCs are neurodegenerative diseases(Checkoway et al., 2011). The current drug
predominantly found in specific regions, notably the subventricular treatments and surgical interventions for neurodegenerative diseases are
zone (SVZ) and the dentate gyrus of the hippocampus(Grochowski et al., unable to effectively halt the degenerative processes(Sudhakar and
2018). The behavior of NSCs is regulated by various factors, determining Richardson, 2019). Nowadays, stem cell therapy offers promising stra­
whether they remain quiescent or enter an active state(Lugert et al., tegies for addressing a wide range of neurodegenerative diseases(Sak­
2010). In the adult human hippocampus, approximately 700 new neu­ thiswary and Raymond, 2012).
rons are generated each day, albeit with a moderate decline associated
with aging(Spalding et al., 2013). These newly formed neurons 2.2.1. Parkinson’s disease
contribute significantly to the plasticity of the nervous system(Toda and PD is a prevalent progressive neurodegenerative disorder charac­
Gage, 2018). The continuous depletion of the neural stem cell (NSC) terized by a deficiency of dopamine (DA) in the striatum, primarily due
pool within the hippocampus is considered the primary cause of age- to the loss of DA-producing neurons located in the substantia nigra
related decline in neurogenesis(Encinas and Sierra, 2012). Both exoge­ (Buddhala et al., 2015). The current primary treatment approach for PD
nous NSC transplantation and endogenous NSC regulation have been involves the administration of levodopa, a precursor of DA, to
extensively investigated in numerous CNS diseases, including Alz­ compensate for the DA deficiency resulting from the destruction of
heimer’s disease (AD) and stroke(Lilja et al., 2015; Huang and Zhang, dopaminergic neurons(Jankovic and Tan, 2020). While drug treatment
2019). Given the robust differentiating capacity of NSCs into neuronal can alleviate symptoms, it does not halt or reverse the progression of
cells, transplantation of NSCs has demonstrated effective promotion of neurodegeneration(Reich and Savitt, 2019).
neural function recovery in preclinical studies(Huang et al., 2021). The transplantation of human embryonic dopaminergic neurons into
the striatum of PD patients holds promise as a potential treatment(Freed
2.1.4. Mesenchymal stem cells et al., 2001). Transplanted stem cells have demonstrated survival in
MSCs possess the ability for self-renewal and multi-directional dif­ patients with severe PD, successfully differentiating into functional DA
ferentiation into various cell types, and they can be derived from nearly neurons and continuing DA production(Piccini et al., 1999). However,
any type of tissue(Ding et al., 2011). Among them, bone marrow-derived the clinical effectiveness of stem cell transplantation remains variable,

2
C. Ying et al. Experimental Neurology 369 (2023) 114543

possibly due to differences among transplanted cells(Olanow et al., accompanied by neuronal death(Sakthiswary and Raymond, 2012).
2003). Despite existing challenges in generating the appropriate type of Stem cells possess the ability to repair damaged neuronal tissue, replace
DA neurons and improving conditions to enhance the survival of damaged or lost cells with differentiated neuronal cells, create a favor­
transplanted cells, there is optimism regarding the potential of stem cells able environment for neural regeneration, or provide protection to
to induce the generation of functional DA neurons and serve as a form of existing healthy neurons and glial cells against further damage
cell therapy(Kim, 2011). (Sivandzade and Cucullo, 2021). While there are currently numerous
clinical trials focused on stem cell therapy for neurodegenerative dis­
2.2.2. Alzheimer’s disease eases, it is important to acknowledge that several challenges and ob­
AD is the most prevalent cause of dementia(Terry and Davies, 1980). stacles must be addressed before the widespread clinical utilization of
The primary pathological mechanism of AD involves the accumulation stem cells(Bednar and Perry, 2012).
of amyloid-β (Aβ) protein, leading to the degeneration and loss of neu­
rons and synapses in various regions of the brain(Moreno-Jiménez et al., 2.3. Stem cell therapies for central nervous system injury
2019; Huang et al., 2021). The cholinergic system plays a crucial role in
the regulation of learning and memory. Hence, the use of acetylcho­ Stem cell therapy is not only applied to neurodegenerative diseases,
linesterase inhibitors (AChEI) can partially alleviate cognitive impair­ but also applied to CNS injury, such as TBI, SCI and stroke(Nout-Lomas,
ment in AD(Musiał et al., 2007). However, as drug therapy for AD is 2022). While the microenvironment of the nervous system in the acute
primarily palliative and does not slow down or reverse the progression stage of injury may not be conducive to the survival and differentiation
of the disease, effective treatment options are needed to address the of NSCs, studies have shown that transplantation of stem cells at specific
needs of AD patients(Park et al., 2020). Stem cell therapy for AD holds time points can be feasible(Ogawa et al., 2002). Stem cell trans­
promise in meeting this requirement. plantation holds promise as an appealing approach to restore nerve
Transplantation of NSCs has shown promise in enhancing endoge­ function in individuals following nerve injuries(Wang et al., 2017).
nous neurogenesis and preventing further cognitive decline, as evi­
denced by animal experiments(Lilja et al., 2015). However, the 2.3.1. Traumatic brain injury
therapeutic potential of NSCs can be hindered by pathological high TBI is a prevalent condition in neurosurgery characterized by a high
levels of oxidative stress, which can damage neurons and impair mortality and disability rate. Despite numerous therapeutic approaches
neurogenic effects(Huang et al., 2021). Current strategies primarily attempted for TBI, many patients with head injuries continue to expe­
focus on directing the differentiation of NSCs into neurons and opti­ rience significant brain damage and behavioral disorders(Wang et al.,
mizing the microenvironmental components to enhance neurogenesis 2012). Currently, there is no clinical treatment available that can reverse
(Yu et al., 2020). Integration of NSC transplantation with gene editing, the pathological cascade of cell death and effectively enhance neuro­
nanomaterials, drug interventions, and other approaches has been behavioral outcomes(Longhi et al., 2005). Promising strategies
explored to improve the efficacy of stem cell therapy(Choi et al., 2018; involving drug-based or cell-based therapies, such as erythropoietin,
Bhattarai et al., 2020; Yu et al., 2020). Nonetheless, transitioning from statins, bone marrow stromal cells, and stem cells, have been explored to
animal experiments to clinical applications still requires significant ef­ promote brain remodeling through processes like angiogenesis, neuro­
forts and research endeavors. genesis, and synaptogenesis, which aim to improve the functional out­
comes for individuals with TBI(Xiong et al., 2009). In recent years, NSC
2.2.3. Amyotrophic lateral sclerosis therapy has garnered significant attention in the field.
ALS is a fatal neurodegenerative disease in adults(Hulisz, 2018). ALS MSCs have demonstrated potential in the treatment of TBI due to
simultaneously accumulate upper motor neurons (UMN) and lower their ability to promote neural regeneration and exert neurotrophic ef­
motor neurons (LMN), which manifested as a progressive paralysis fects(Wang et al., 2017). Human amnion-derived mesenchymal stem
(Ustyantseva et al., 2020). The disease typically progresses rapidly, with cells (AMSCs) and BM-MSCs are promising sources of MSCs that have
respiratory failure and death occurring within one to five years after shown the ability to enhance the expression of neurotrophic factors,
symptom onset(Yang et al., 2021). While the exact pathogenesis of ALS thereby facilitating nerve regeneration(Guan et al., 2013; Anbari et al.,
remains unclear, neuronal dysfunction is believed to be caused by ge­ 2014). However, stem cell therapy still faces several challenges,
netic mutations and abnormal protein deposition(Oskarsson et al., including the low survival rate of transplanted stem cells and their
2018). Among the various drugs tried for ALS patients, riluzole is limited differentiation capacity(Bae et al., 2021). Combining stem cell
currently the only approved medication that directly modifies the course therapy with biomaterials holds promise for addressing these challenges
of the disease(Dharmadasa and Kiernan, 2018). Numerous therapies, by improving the survival rate of stem cells, creating a conducive
including anti-apoptotic drugs, anti-oxidant drugs, anti-aggregation environment for their survival, and supporting the regeneration of TBI
drugs, and anti-excitotoxicity drugs, are being investigated at various (Liu et al., 2022).
stages of preclinical and clinical evaluation(Nowicka et al., 2019).
However, single-drug treatments have not been able to reverse the loss 2.3.2. Spinal cord injury
of motor neurons (MNs) seen in ALS(Ralli et al., 2019). SCI is a severe condition characterized by the abrupt loss of sensory,
With the expanding research on stem cells, particularly iPSCs, stem motor, and autonomic functions below the level of the injury(Mothe and
cell therapy has emerged as a promising approach for MNs regeneration Tator, 2012). Despite extensive efforts and resources invested, no
(Kim et al., 2014). In comparison to MSCs, NSCs derived from the spinal treatment has been able to achieve substantial improvement or signifi­
cord not only release beneficial growth factors but also integrate into the cant functional recovery of the injured nerves(Tator, 2006). Current
neural circuitry of the MNs and spinal cord, forming appropriate syn­ treatment approaches primarily focus on reducing secondary compli­
apses(Lunn et al., 2014). Although the safety of stem cell transplantation cations and maximizing residual function through rehabilitation efforts
in ALS patients has been demonstrated, there are still several challenges (Dell’Anno and Strittmatter, 2017). The differentiation of NSCs within
that need to be addressed in this treatment approach, including the the spinal cord holds promise as a potential strategy to replenish the lost
handling of stem cells and monitoring clinical outcomes(Giacomelli neurons following SCI(Zhao et al., 2017a).
et al., 2022). Several types of NSCs, including MSCs derived from various tissue
In addition to the mentioned neurodegenerative diseases, there are sources, ESCs, and iPSCs, have been investigated for their potential in
various types of neurodegenerative diseases. The fundamental mecha­ treating SCI(Shao et al., 2019). Endogenous NSCs within the spinal cord
nism underlying most neurodegenerative diseases involves the pro­ are also considered as therapeutic targets for SCI, although their func­
gressive loss of neuronal structure, function, or number, often tion requires external regulation(Gilbert et al., 2022). When NSC grafts

3
C. Ying et al. Experimental Neurology 369 (2023) 114543

are transplanted into the site of SCI, they can integrate and form func­ promoting neural regeneration(Ford et al., 2020).
tional synaptic subnetworks, which can be beneficial for the recovery of
nerve function(Ceto et al., 2020). Furthermore, NSC-derived exosomes 3.1.1. Neurotrophic effects
and small extracellular vesicles have been identified as important me­ Stem cell therapy offers neuroprotection through the secretion of
diators in promoting nerve regeneration(Zhong et al., 2020). Enhancing various growth factors, such as nerve growth factor (NGF), brain-
the delivery of NSCs can be achieved through approaches such as stent derived neurotrophic factor (BDNF), glial cell line-derived neuro­
placement or combined immunotherapy, which have shown potential in trophic factor (GDNF) and neurotrophin-3 (NT-3) (Blesch et al., 2002;
improving the treatment efficacy for SCI(Liau et al., 2020; Chen et al., Oliveira et al., 2013). The ability of stem cells to provide sustained
2022). release of these growth factors is a significant advantage compared to
direct injection, as growth factors administered directly can be rapidly
2.3.3. Stroke diluted and have short half-lives(Ritfeld et al., 2011). Furthermore,
Stroke, particularly ischemic stroke, is a leading cause of mortality these growth factors not only enhance the neuroprotective effects of
and disability globally, with ischemic stroke accounting for over 80% of stem cell therapy but also promote the proliferation and differentiation
all stroke cases(Benjamin et al., 2017). While reperfusion therapy is of stem cells, thereby reinforcing the overall nerve regeneration poten­
currently the main effective treatment, its administration is time- tial(Jin-qiao et al., 2009) (Fig. 1).
sensitive, and only a limited number of patients can benefit from it NGF promotes the proliferation of stem cells and inhibit their dif­
(Hankey, 2017). Stroke-related disabilities often lead to long-term im­ ferentiation into neurons by activating the phosphorylation of Erk1/2
pairments, and conventional rehabilitation approaches often yield (Wang et al., 2009). However, some studies have shown that NGF pro­
limited results, imposing significant social and economic burdens(Jiao motes the differentiation of stem cells into mature neural phenotype,
et al., 2021). Stem cell transplantation holds great promise as a potential which may be determined by the concentration of NGF(Levenberg et al.,
treatment for stroke, and extensive research has been conducted in this 2005).
area(Huo et al., 2014; Zhao et al., 2022). Multiple studies have demonstrated that the combination of stem
Stem cell therapy is expected to reconstruct the neural network cells and BDNF promotes axonal extension(Bhattarai et al., 2020; Li
which was injured by stroke(Kalluri and Dempsey, 2008). NSC trans­ et al., 2023). The protective effect of BDNF on apoptosis induction in
plantation has been proved to promote functional recovery after brain stem cells is thought to be mediated through the PI3K/Akt and MAPK
injury through a variety of by stander mechanisms, rather than simple signaling pathways via Trk receptors(Nguyen et al., 2009).
cell replacement(Huang and Zhang, 2019). NSCs exhibit anti- GDNF has demonstrated neuroprotective effects in animal models of
inflammatory properties in stroke treatment, exerting their effects various CNS diseases(Shi et al., 2009; Dulz et al., 2020). The concurrent
through mechanisms such as anti-apoptosis, angiogenesis, production of administration of GDNF during the early stage of stem cell trans­
growth factors, neuroprotection, and anti-fibrosis(Trounson and plantation improves the stem cell microenvironment, enhances stem cell
McDonald, 2015). Furthermore, intra-venous and intra-arterial admin­ survival, and enhances therapeutic efficacy(Zhao et al., 2017b). The
istration of BM-MSCs and MSCs have also shown effectiveness in stroke specific mechanisms underlying GDNF-induced neurogenesis and neu­
treatment, mediated by neuroprotective mechanisms(Chrostek et al., roprotection have not been fully elucidated, but they are believed to
2019). Therefore, further research exploring the role and mechanisms of involve the PI3K/Akt and MAPK signaling pathways(Nicole et al.,
stem cells in nerve regeneration following stroke holds significant 2001).
importance for future clinical applications. NT-3 shares structural and functional similarities with NGF and
In addition, stem cell therapy is also used for peripheral nerve injury, BDNF, and it also plays a role in promoting nerve repair in stem cells(Wu
ischemic hypoxic encephalopathy, and other nerve injury diseases, et al., 2018; Sun et al., 2020; Li et al., 2021). The impact of NT-3 on cell
which plays a role in nerve regeneration and anti-inflammatory, and proliferation is dose-dependent, with low doses promoting proliferation
improve nerve damage(Shi et al., 2009; Feng et al., 2013; Donega et al., and high doses reducing proliferation and inducing stem cell differen­
2014; Lee et al., 2016; Saeki et al., 2022). Further research and clinical tiation(Hapner et al., 2006).
trials are still needed if stem cell therapy is to be used in clinical The influence of neurotrophic factors on stem cells contributes to the
treatment. reinforcement of NSC therapy. Enhancing the expression of neuro­
trophic factors in stem cells or combining them with biomaterials that
3. Molecular mechanisms underlying stem cell therapies for release neurotrophic factors at appropriate doses can be advantageous
central nervous system diseases (Li et al., 2021).

In recent years, significant progress has been made in understanding 3.1.2. Immunoregulatory effect
the pathophysiological mechanisms of various neurological diseases, In CNS diseases, particularly acute injury conditions, inflammatory
which has identified numerous potential targets for stem cell therapy. In reactions play a significant role in the regeneration of the nervous sys­
this part, we will review the molecular mechanism of stem cell therapies tem(Gaudet and Fonken, 2018; Ghaddar et al., 2021; Huang et al.,
for CNS diseases from both preclinical evidence and clinical findings. 2022). Thus, reducing the detrimental effects of inflammatory factors
and creating a microenvironment that promotes neuronal regeneration
3.1. Preclinical evidence of stem cell therapies for central nervous system is an effective strategy in stem cell therapy(Gao et al., 2022) (Fig. 2).
diseases In a variety of CNS diseases, inflammation is an important factor
causing the loss of nerve function, and is also the target of multiple drug
Preclinical research has provided valuable insights into the molec­ treatments(Dooley et al., 2014). For instance, in many animal models,
ular mechanisms underlying stem cell-based neural regeneration ther­ TNF-α and IFN-γ have been found to inhibit the proliferation of stem
apy. Stem cells have been found to secrete growth factors and cytokines cells(Ben-Hur et al., 2003). Anti-inflammatory drugs have shown sig­
that facilitate neural survival, axon regeneration, and neurogenesis, nificant efficacy in promoting nerve regeneration by facilitating the
contributing to the recovery of neural function(De Gioia et al., 2020). clearance of reactive oxygen species (ROS) and reducing the secretion of
Moreover, stem cells exhibit immunomodulatory properties, reducing inflammatory factors by macrophages(Gao et al., 2022).
inflammation and modulating the immune response, which can have a Stem cells have demonstrated a positive response to inflammation, as
positive impact on neural regeneration(Xian et al., 2019). Additionally, evidenced by studies highlighting the essential role of inflammation in
the integration of stem cells into neural tissue and their ability to form facilitating stem cell migration to injury sites(Kizil et al., 2015). In vitro
functional connections with host neurons play a crucial role in investigations have further revealed that inflammatory signals, such as

4
C. Ying et al. Experimental Neurology 369 (2023) 114543

Fig. 1. Neurotrophic effects of stem cells. Stem cells exert neuroprotective effects through the secretion of various growth factors, thereby enhancing their overall
neuroprotective potential. These growth factors have the capacity to influence different types of stem cells, regulating their proliferation and differentiation through
distinct signaling pathways. Simultaneously, these growth factors can also interact with the neural microenvironment, mediating neuroprotective effects by acti­
vating Trk receptors.

TNF-α or IL-1β, can stimulate stem cell proliferation via the activation of recovery in experimental animals often occurs earlier than the time
NF-κB and JNK signaling pathways(Wang et al., 2007). Notably, TNF-α required for the functional integration of transplanted stem cells, which
knockout mice do not manifest the behavioral recovery observed in may be attributed to the rapid and highly spontaneous functional re­
wild-type mice following TBI(Scherbel et al., 1999). Intriguingly, the covery observed in most experimental animals(Baker et al., 2019).
downregulation of the anti-inflammatory factor IL-10 actually promotes Nonetheless, it is widely believed that the long-term and effective cell
the differentiation of neural progenitor cells(Perez-Asensio et al., 2013). replacement achieved through stem cell therapy plays a crucial role in
Additionally, in zebrafish, inflammation has been identified as an active the overall therapeutic outcome(Tornero et al., 2013).
regulator of CNS neuron regeneration(Kyritsis et al., 2012). Simply Furthermore, it is possible to induce transplanted stem cells to
inhibiting inflammatory reaction does not seem to be the best way to differentiate into specific subtypes of neurons, thereby enhancing the
promote nerve regeneration. therapeutic effects. For instance, inducing stem cells to differentiate into
In conclusion, stem cell therapy for nerve regeneration should dopaminergic neurons and transplanting them into Parkinson’s animal
consider the timing of intervention, the specific cytokines used, and the model enhance the therapeutic effect(Hargus et al., 2010).
regulatory scheme, as different cytokines can have varying effects on Stem cells provide a unique opportunity to reduce neurological
NSCs at different times(Mueller et al., 2005). damage through multimodal treatment. Numerous preclinical studies
have demonstrated that stem cells exhibit neuroprotective effects by
3.1.3. Cell replacement mitigating secondary injury through anti-inflammatory mechanisms,
In addition to the various mechanisms mediated by neurotrophic facilitating endogenous neurogenesis and synaptic remodeling, and
factor signaling pathways to protect and promote nerve recovery, stem acting as cell substitutes to promote tissue and functional recovery
cells can also serve as cell substitutes themselves(Jiao et al., 2021) (Baker et al., 2019). Additionally, stem cell therapy promotes neural
(Fig. 3). function recovery by stimulating angiogenesis, facilitating blood-brain
Some studies have demonstrated that following injury, a significant barrier repair, and enhancing the formation of neural synapses(Kana­
influx of stem cells occurs at the injury site, where they undergo dif­ zawa et al., 2017). These preclinical findings lay the groundwork for
ferentiation into neurons, thereby inhibiting glial scar formation and cell further clinical research in this field.
death(Song et al., 2015). Various markers indicative of neural differ­
entiation stages, such as Nestin, Tuj1, NeuN, TH, DARPP-32, and SV38, 3.2. Clinical findings of limitations of stem cell therapies for central
have been detected in these migrating cells(Chang et al., 2013). Trans­ nervous system diseases
planted cells have been observed to differentiate into diverse neuronal
subtypes expressing GABA, parvalbumin, calretinin, or calbindin(Oki While stem cell therapy has demonstrated significant advantages in
et al., 2012). preclinical research, progress in clinical studies has been somewhat
However, previous studies have indicated that the neural function limited (Table.1). Stem cell therapy, whether delivered through arterial

5
C. Ying et al. Experimental Neurology 369 (2023) 114543

Fig. 2. The interaction between inflammatory immunity and stem cells. On the one hand, inflammation exerts a dual regulatory effect on stem cells. Various
chemokines and inflammatory mediators can modulate the migration, proliferation, and differentiation of stem cells. Additionally, the presence of reactive oxygen
species can induce apoptosis in both stem cells and neurons. On the other hand, stem cells themselves can influence different types of immune cells through various
mediators, either promoting or inhibiting the inflammatory response.

or intravenous injection or targeted transplantation, has been proved to stem cell transplantation, further research is needed to determine
be safe by many clinical studies, but has little effect(Trounson and optimal parameters, including cell type, cell dosage, and injection
McDonald, 2015). method.
Stem cell therapy holds great promise in the treatment of neurode­
generative diseases such as AD, where specific effective drugs are 4. Perspectives and challenges in stem cell therapies for central
currently lacking(Checkoway et al., 2011). Although preclinical studies nervous system diseases
have studied the source, differentiation efficiency, culture method,
tumor formation, injection route and mobility of stem cells, and ach­ 4.1. Emerging trends in stem cell research for central nervous system
ieved satisfactory results, no significant clinical effects have been diseases
observed in several clinical studies(Kang et al., 2016). Several factors,
including treatment timing, patient heterogeneity, and loss to follow-up, Based on the research on the mechanism of stem cell therapy and the
can influence the outcomes of clinical research(Liu et al., 2020). More outcomes of clinical trials, it appears challenging to solely rely on stem
clinical trials are in progress, and further steps are needed before stem cell transplantation for direct promotion of nerve regeneration. There
cell therapy becomes a clinically feasible method to treat related are emerging trends in research related to stem cell therapy that warrant
diseases. discussion.
In the clinical research of nerve injury, stem cell therapy has not
yielded satisfactory results. While stem cell transplantation has gained 4.1.1. Stem cell acquisition
attention as a potential treatment for stroke, its clinical application re­ The acquisition of stem cells has been a prominent research focus in
mains significantly limited(Zhao et al., 2022). Huo et al. conducted a stem cell therapy, especially with the emergence of iPSCs and induced
comprehensive analysis of clinical trials focused on stem cell trans­ neural progenitor cells (iNPCs). iPSCs, derived from adult cells through
plantation in stroke, and they concluded that there is currently insuffi­ genetic reprogramming to exhibit properties akin to ESCs, possess the
cient evidence to support the replacement of damaged cells, capacity for differentiation into various cell types, including neurons.
reconstruction of neural circuits, or improvement of functional loss (Aboul-Soud et al., 2021). iNPCs can be reprogrammed via astrocytes
through stem cell transplantation after stroke(Huo et al., 2014). Fortu­ through endothelial cell regulation or through the overexpression of
nately, most of the methods employed for stem cell transplantation transcription factors, with different differentiation potential for
appear to be safe(Kawabori et al., 2020). To enhance the effectiveness of neuronal subtypes(Ma et al., 2018; Li et al., 2022). Researchers are

6
C. Ying et al. Experimental Neurology 369 (2023) 114543

Fig. 3. The cell replacement effect of stem cell therapy. In stem cell therapy for the central nervous system, various types of stem cells can express various neuronal
markers through pathways such as “migrate/transplantion” or “induced differentiation”, contributing to their role in cell replacement.

actively investigating the potential of induced pluripotent cells for 4.1.4. Combination therapies
neural regeneration, as they can be derived from a patient’s own cells, Researchers are actively investigating combination therapies that
minimizing the risk of immune rejection(Du et al., 2022). combine stem cells with other modalities, including drugs and physical
therapy, to enhance treatment outcomes(Lukovic et al., 2015). For
4.1.2. Gene editing instance, the combination of stem cell transplantation with rehabilita­
Gene editing has emerged as a valuable tool in stem cell therapy with tion therapy or electromagnetic stimulation has shown promise in pro­
several applications. Firstly, it plays a crucial role in the conversion of moting the repair of SCI(Zheng et al., 2020; Lu et al., 2022).
neurons into iPSCs(Tazaki et al., 2017). Gene editing techniques,
including CRISPR/Cas9, are employed to modify stem cells, enhancing 4.1.5. Anti immune rejection
their potential for neural regeneration(Yang et al., 2022). Secondly, Due to the immune rejection in stem cell transplantation, researchers
researchers are utilizing gene editing to rectify genetic mutations asso­ are attempting to reduce its adverse effects from multiple perspectives.
ciated with neurological disorders, such as Huntington’s disease(Golas MSCs inhibit T and B cell activity and can be used to suppress immune
and Sander, 2016). Furthermore, gene editing can empower stem cells to mediated diseases and transplant rejection reactions(Uccelli et al.,
secrete specific neurotrophic factors like BDNF, thereby promoting 2007). Compared to NSCs transplantation, MSCs transplantation elicits
neuronal regeneration(Deng et al., 2016). weaker immune responses and provides long-term behavioral benefits
(Rossignol et al., 2014). Additionally, biomaterials such as hydrogels are
4.1.3. 3D bioprinting being investigated to modulate the immune microenvironment post-
3D bioprinting technology is being employed to fabricate brain-like implantation, enhancing the survival and functionality of stem cells,
structures, mimicking the intricate complexity of the human brain. By and working in conjunction with immunosuppressive drugs to regulate
utilizing different types of 3D biomaterials, such as scaffolds, the neural immune rejection(Gao et al., 2023).
microenvironment surrounding stem cells can be regulated, effectively Overall, the emerging trends in stem cell research for neural regen­
guiding their growth and differentiation(Koffler et al., 2019). Integra­ eration show great promise in the development of novel treatments for
tion of stem cell transplantation with 3D biomaterials holds promise in neurological disorders and injuries. However, it is important to
promoting nerve regeneration and augmenting the efficacy of stem cell emphasize the need for further research to comprehensively evaluate
therapy(Chen et al., 2022). the safety and efficacy of these therapies. Continued investigation and
rigorous clinical trials will be essential to ensure the successful

7
C. Ying et al. Experimental Neurology 369 (2023) 114543

Table 1 cells(Gao et al., 2023). To mitigate the risk of immune rejection, re­
Summary of stem cells administered in clinical trials in the last 10 years. searchers are actively investigating diverse strategies, including the
Disease Stem Routes No. Outcome Reference utilization of patient-specific stem cells and immune suppression ther­
Target cell cases apy(Rosenzweig et al., 2018). While there is a foundation of research in
types the field of immune rejection, further studies are warranted to enhance
AD hUCB- Stereotactic 9/9 No adverse (Kim et al., its application in the context of stem cell therapy(Uccelli et al., 2007;
MSCs brain injection events; 2015) Rossignol et al., 2014; Gao et al., 2023).
No dose-
limiting
toxicity;
4.2.2. Tumor formation
No Tumorigenicity and pluripotent differentiation potential are coupled
improvement cell properties unified by neural stemness(Zhang et al., 2022). There­
ALS MSCs Intrathecal 19/ No adverse (Petrou fore, there is a risk of tumor formation associated with stem cell thera­
injection 19 events; et al.,
pies(Johnson et al., 2008; Ritfeld et al., 2011). Although some clinical
Some patients 2021)
had a > 25% trials have not found that transplanted NSCs are associated with tumor
improvement formation, more clinical research data are needed to assess the safety of
IS AD- Intravenous 4/13 No adverse (de Celis- treatment before large-scale clinical application(Yan et al., 2013; Chang
MSCs injection events; Ruiz et al., et al., 2013; Lee et al., 2016).
No tumor 2022)
formation;
No 4.2.3. Regulatory approval
improvement Stem cell therapies for neural regeneration encounter significant
ALS hNSCs Microinjections 18/ No adverse (Mazzini regulatory challenges pertaining to safety, efficacy, and ethical aspects
into the gray 18 events; et al.,
(Sonntag et al., 2018). The regulatory approval process is multifaceted,
matter tracts No tumor 2019)
formation; necessitating comprehensive preclinical and clinical investigations
Limited while ensuring adherence to ethical standards and regulatory
efficacy guidelines.
IS ABMD- Intravenous 16/ No adverse (Jaillard In summary, the resolution of these technical challenges is of utmost
MSCs injection 31 events; et al.,
importance in the advancement of safe and effective stem cell therapies
No 2020)
improvement for nerve regeneration. While there is still much work to be done,
in neurological ongoing research endeavors are making significant strides in tackling
function score; these obstacles and bringing stem cell therapies closer to real-world
Improvement
clinical applications.
in motor
function score
SCI AD- Intrathecal 1/1 No adverse (Bydon 4.3. Ethical considerations for stem cell therapies
MSCs injection events; et al.,
Meaningful 2020) The clinical application of stem cell therapy requires serious
improvement
SCI hiPSC- Intrathecal 4/4 Not yet (Sugai
consideration of ethical issues. The ethical considerations mainly
NS/PC injection completed; et al., include the following aspects.
To evaluate 2021) Source of stem cells: The source of stem cells is a significant ethical
safety consideration. ESCs, which are derived from human embryos, are highly
IS hNSCs Stereotactic 11/ No adverse (Kalladka
pluripotent and have the potential to develop into any cell type in the
ipsilateral 13 events; et al.,
putamen Decreased 2016) body. However, the use of ESCs raises ethical concerns related to the
injection infarct destruction of human embryos(Sugaya and Vaidya, 2018). The research
of iPSCs does not involve corresponding ethical issues, which is a po­
Abbreviations: No. cases: number of cases in the treatment group/total number
of cases; hUCB-MSCs: human umbilical cord blood-derived mesenchymal stem
tential alternative(Du et al., 2022).
cells; AD-MSCs: adipose tissue-derived mesenchymal stem cells. Informed consent: Informed consent is an essential ethical consid­
IS: Ischemic Stroke; hNSCs: human neural stem cells; ABMD-MSCs: autologous eration for any medical procedure, including stem cell therapies. Pa­
bone marrow-derived mesenchymal stem cells; hiPSC-NS/PC: induced pluripo­ tients must be fully informed of the risks and benefits of stem cell
tent cell-derived neural stem/progenitor cell. therapies and must provide informed consent before treatment.
Fairness and access: The availability and affordability of stem cell
translation of these advancements into effective and reliable treatments therapies can raise concerns about fairness and access. Stem cell thera­
for patients. pies are often expensive and may not be accessible to all patients,
leading to potential disparities in access to treatment.
In summary, careful consideration of ethical issues is crucial to
4.2. Technical challenges in stem cell therapies for central nervous system ensuring that stem cell therapies are developed and used in a responsible
diseases and ethical manner.

Based on the results of current clinical research, although stem cell 5. Conclusion
therapies hold promise for neural regeneration, there are several tech­
nical challenges that need to be addressed before these therapies can be Stem cell therapies for neural regeneration hold tremendous poten­
widely used in clinical settings(Chang et al., 2020). tial in the treatment of CNS diseases and injuries. Extensive preclinical
and clinical studies have utilized various types of stem cells, each with
4.2.1. Immune rejection their own advantages and limitations. These therapies have shown
Immune rejection poses a significant challenge in stem cell trans­ promise in addressing conditions like PD, stroke, and SCI, shedding light
plantation treatments, affecting their overall efficacy and success on the underlying molecular mechanisms of neural regeneration.
(Sackett et al., 2016). The transplantation of stem cells can activate However, technical limitations, ethical considerations, and regulatory
immune responses that may result in the rejection of the transplanted challenges still need to be addressed to advance this field further.

8
C. Ying et al. Experimental Neurology 369 (2023) 114543

Encouragingly, emerging trends in gene editing and tissue engineering Benjamin, E.J., Blaha, M.J., Chiuve, S.E., Cushman, M., Das, S.R., Deo, R., de Ferranti, S.
D., Floyd, J., Fornage, M., Gillespie, C., Isasi, C.R., Jiménez, M.C., Jordan, L.C.,
present opportunities for overcoming these obstacles. Ongoing research
Judd, S.E., et al., 2017. Heart disease and stroke statistics-2017 update: a report from
is essential to enhance the safety and efficacy of stem cell therapies while the American Heart Association. Circulation 135 (10), e146–e603.
ensuring a thoughtful exploration of their ethical implications. Bhattarai, P., Cosacak, M.I., Mashkaryan, Demir, S., Popova, S.D., Govindarajan, N.,
Brandt, K., Zhang, Y., Chang, W., Ampatzis, K., Kizil, C., 2020. Neuron-glia
interaction through Serotonin-BDNF-NGFR axis enables regenerative neurogenesis
Ethics approval and consent to participate in Alzheimer’s model of adult zebrafish brain. PLoS Biol. 18 (1) e3000585.
Bifari, F., Decimo, I., Pino, A., Llorens-Bobadilla, E., Zhao, S., Lange, C., Panuccio, G.,
Boeckx, B., Thienpont, B., Vinckier, S., Wyns, S., Bouché, A., Lambrechts, D.,
Not applicable.
Giugliano, M., et al., 2017. Neurogenic radial glia-like cells in meninges migrate and
differentiate into functionally integrated neurons in the neonatal cortex. Cell Stem
Consent for publication Cell 20 (3), 360–373.e7.
Blesch, A., Lu, P., Tuszynski, M.H., 2002. Neurotrophic factors, gene therapy, and neural
stem cells for spinal cord repair. Brain Res. Bull. 57 (6), 833–838.
Not applicable. Buddhala, C., Loftin, S.K., Kuley, B.M., Cairns, N.J., Campbell, M.C., Perlmutter, J.S.,
Kotzbauer, P.T., 2015. Dopaminergic, serotonergic, and noradrenergic deficits in
Parkinson disease. Ann. Clin. Translat. Neurol. 2 (10), 949–959.
Availability of data and materials Bydon, M., Dietz, A.B., Goncalves, S., Moinuddin, F.M., Alvi, M.A., Goyal, A., Yolcu, Y.,
Hunt, C.L., Garlanger, K.L., Del Fabro, A.S., Reeves, R.K., Terzic, A., Windebank, A.
Not applicable. J., Qu, W., 2020. CELLTOP clinical trial: first report from a phase 1 trial of
autologous adipose tissue-derived mesenchymal stem cells in the treatment of
paralysis due to traumatic spinal cord injury. Mayo Clin. Proc. 95 (2), 406–414.
Funding Ceto, S., Sekiguchi, K.J., Takashima, Y., Nimmerjahn, A., Tuszynski, M.H., 2020. Neural
stem cell grafts form extensive synaptic networks that integrate with host circuits
after spinal cord injury. Cell Stem Cell 27 (3), 430–440.e5.
This study was supported by the National Natural Science Founda­ Chang, D.-J., Oh, S.-H., Lee, N., Choi, C., Jeon, I., Kim, H.S., Shin, D.A., Lee, S.E., Kim, D.,
tion of China (81870964) and Zhejiang Provincial Natural Science Song, J., 2013. Contralaterally transplanted human embryonic stem cell-derived
Foundation of China (LY23H090012). The funding body played no role neural precursor cells (ENStem-A) migrate and improve brain functions in stroke-
damaged rats. Exp. Mol. Med. 45 (11) e53.
in the design of the study and collection, analysis, and interpretation of Chang, C.-Y., Ting, H.-C., Liu, C.-A., Su, H.-L., Chiou, T.-W., Lin, S.-Z., Harn, H.-J., Ho, T.-
data and in writing the manuscript. J., 2020. Induced pluripotent stem cell (iPSC)-based neurodegenerative disease
models for phenotype recapitulation and drug screening. Molecules (Basel,
Switzerland) 25 (8).
Authors’ contributions Checkoway, H., Lundin, J.I., Kelada, S.N., 2011. Neurodegenerative diseases. IARC Sci.
Publ. 163, 407–419.
CY and JZ contributed to literature collection and drafting the Chen, P., Xu, C., Wu, P., Liu, K., Chen, F., Chen, Y., Dai, H., Luo, Z., 2022. Wirelessly
powered electrical-stimulation based on biodegradable 3D piezoelectric scaffolds
manuscript. HZ contributed to manuscript and figures revisions during promotes the spinal cord injury repair. ACS Nano 16 (10), 16513–16528.
the revision work period. SG contributed to design of figures and tables. Choi, S.H., Bylykbashi, E., Chatila, Z.K., Lee, S.W., Pulli, B., Clemenson, G.D., Kim, E.,
XG and JL collected the literatures. YH and HW designed the study and Rompala, A., Oram, M.K., Asselin, C., Aronson, J., Zhang, C., Miller, S.J.,
Lesinski, A., et al., 2018. Combined adult neurogenesis and BDNF mimic exercise
critically revised the manuscript. All authors contributed to the article effects on cognition in an Alzheimer’s mouse model. Science (New York, N.Y.) 361
and approved the submitted version. (6406).
Chrostek, M.R., Fellows, E.G., Crane, A.T., Grande, A.W., Low, W.C., 2019. Efficacy of
stem cell-based therapies for stroke. Brain Res. 1722, 146362.
Declaration of Competing Interest Đặng, T.C., Ishii, Y., Nguyen, V.D., Yamamoto, S., Hamashima, T., Okuno, N., Nguyen, Q.
L., Sang, Y., Ohkawa, N., Saitoh, Y., Shehata, M., Takakura, N., Fujimori, T.,
Inokuchi, K., et al., 2019. Powerful homeostatic control of oligodendroglial lineage
The authors declare that they have no competing interests. by PDGFRα in adult brain. Cell Rep. 27 (4), 1073–1089.e5.
de Celis-Ruiz, E., Fuentes, B., Alonso de Leciñana, M., Gutiérrez-Fernández, M.,
Borobia, A.M., Gutiérrez-Zúñiga, R., Ruiz-Ares, G., Otero-Ortega, L., Laso-García, F.,
Data availability
Gómez-de Frutos, M.C., Díez-Tejedor, E., 2022. Final results of allogeneic adipose
tissue-derived mesenchymal stem cells in acute ischemic stroke (AMASCIS): A phase
No data was used for the research described in the article. II, randomized, double-blind, placebo-controlled, single-center, pilot clinical trial.
Cell Transplant. 31, 9636897221083864.
De Gioia, R., Biella, F., Citterio, G., Rizzo, F., Abati, E., Nizzardo, M., Bresolin, N.,
Acknowledgements Comi, G.P., Corti, S., 2020. Neural stem cell transplantation for neurodegenerative
diseases. Int. J. Mol. Sci. 21 (9).
Decimo, I., Bifari, F., Rodriguez, F.J., Malpeli, G., Dolci, S., Lavarini, V., Pretto, S.,
Not Applicable.
Vasquez, S., Sciancalepore, M., Montalbano, A., Berton, V., Krampera, M.,
Fumagalli, G., 2011. Nestin- and doublecortin-positive cells reside in adult spinal
References cord meninges and participate in injury-induced parenchymal reaction. Stem Cells
(Dayton, Ohio) 29 (12), 2062–2076.
Dell’Anno, M.T., Strittmatter, S.M., 2017. Rewiring the spinal cord: direct and indirect
Aboul-Soud, M.A.M., Alzahrani, A.J., Mahmoud, A., 2021. Induced pluripotent stem cells
strategies. Neurosci. Lett. 652, 25–34.
(iPSCs)-roles in regenerative therapies, disease modelling and drug screening. Cells
Deng, P., Torrest, A., Pollock, K., Dahlenburg, H., Annett, G., Nolta, J.A., Fink, K.D.,
10 (9).
2016. Clinical trial perspective for adult and juvenile Huntington’s disease using
Alexanian, A.R., Maiman, D.J., Kurpad, S.N., Gennarelli, T.A., 2008. In vitro and in vivo
genetically-engineered mesenchymal stem cells. Neural Regen. Res. 11 (5), 702–705.
characterization of neurally modified mesenchymal stem cells induced by epigenetic
Dharmadasa, T., Kiernan, M.C., 2018. Riluzole, disease stage and survival in ALS. Lancet.
modifiers and neural stem cell environment. Stem Cells Dev. 17 (6), 1123–1130.
Neurol. 17 (5), 385–386.
Anbari, F., Khalili, M.A., Bahrami, A.R., Khoradmehr, A., Sadeghian, F., Fesahat, F.,
Ding, D.-C., Shyu, W.-C., Lin, S.-Z., 2011. Mesenchymal stem cells. Cell Transplant. 20
Nabi, A., 2014. Intravenous transplantation of bone marrow mesenchymal stem cells
(1), 5–14.
promotes neural regeneration after traumatic brain injury. Neural Regen. Res. 9 (9),
Donega, V., Nijboer, C.H., Braccioli, L., Slaper-Cortenbach, I., Kavelaars, A., van Bel, F.,
919–923.
Heijnen, C.J., 2014. Intranasal administration of human MSC for ischemic brain
Bae, M., Hwang, D.W., Ko, M.K., Jin, Y., Shin, W.J., Park, W., Chae, S., Lee, H.J., Jang, J.,
injury in the mouse: in vitro and in vivo neuroregenerative functions. PLoS One 9
Yi, H.-G., Lee, D.S., Cho, D.-W., 2021. Neural stem cell delivery using brain-derived
(11), e112339.
tissue-specific bioink for recovering from traumatic brain injury. Biofabrication 13
Dooley, D., Vidal, P., Hendrix, S., 2014. Immunopharmacological intervention for
(4).
successful neural stem cell therapy: new perspectives in CNS neurogenesis and
Baker, E.W., Kinder, H.A., West, F.D., 2019. Neural stem cell therapy for stroke: a
repair. Pharmacol. Ther. 141 (1), 21–31.
multimechanistic approach to restoring neurological function. Brain and behavior 9
Drury-Stewart, D., Song, M., Mohamad, O., Guo, Y., Gu, X., Chen, D., Wei, L., 2013.
(3), e01214.
Highly efficient differentiation of neural precursors from human embryonic stem
Bednar, M.M., Perry, A., 2012. Neurorestoration therapeutics for neurodegenerative and
cells and benefits of transplantation after ischemic stroke in mice. Stem Cell Res Ther
psychiatric disease. Neurol. Res. 34 (2), 129–142.
4 (4), 93.
Ben-Hur, T., Ben-Menachem, O., Furer, Einstein, O., Mizrachi-Kol, R., Grigoriadis, N.,
2003. Effects of proinflammatory cytokines on the growth, fate, and motility of
multipotential neural precursor cells. Mol. Cell. Neurosci. 24 (3), 623–631.

9
C. Ying et al. Experimental Neurology 369 (2023) 114543

Du, X., Amponsah, A.E., Kong, D., He, J., Ma, Z., Ma, J., Cui, H., 2022. hiPSC-neural Jaillard, A., Hommel, M., Moisan, A., Zeffiro, T.A., Favre-Wiki, I.M., Barbieux-
stem/progenitor cell transplantation therapy for spinal cord injury. Curr. Stem Cell Guillot, M., Vadot, W., Marcel, S., Lamalle, L., Grand, S., Detante, O., 2020.
Res. Ther. 18 (4), 487–498. Autologous mesenchymal stem cells improve motor recovery in subacute ischemic
Dugger, B.N., Dickson, D.W., 2017. Pathology of neurodegenerative diseases. Cold Spring stroke: a randomized clinical trial. Transl. Stroke Res. 11 (5), 910–923.
Harb. Perspect. Biol. 9 (7). Jankovic, J., Tan, E.K., 2020. Parkinson’s disease: etiopathogenesis and treatment.
Dulz, S., Bassal, M., Flachsbarth, K., Riecken, K., Fehse, B., Schlichting, S., Bartsch, S., J. Neurol. Neurosurg. Psychiatry 91 (8), 795–808.
Bartsch, U., 2020. Intravitreal co-administration of GDNF and CNTF confers Jiao, Y., Liu, Y.-W., Chen, W.-G., Liu, J., 2021. Neuroregeneration and functional
synergistic and long-lasting protection against injury-induced cell death of retinal recovery after stroke: advancing neural stem cell therapy toward clinical application.
ganglion cells in mice. Cells 9 (9). Neural Regen. Res. 16 (1), 80–92.
Encinas, J.M., Sierra, A., 2012. Neural stem cell deforestation as the main force driving Jin-qiao, S., Bin, S., Wen-hao, Z., Yi, Y., 2009. Basic fibroblast growth factor stimulates
the age-related decline in adult hippocampal neurogenesis. Behav. Brain Res. 227 the proliferation and differentiation of neural stem cells in neonatal rats after
(2), 433–439. ischemic brain injury. Brain and Development 31 (5), 331–340.
Feigin, V.L., Vos, T., Alahdab, F., Amit, A.M.L., Bärnighausen, T.W., Beghi, E., Johnson, T.S., O’Neill, A.C., Motarjem, P.M., Nazzal, J., Randolph, M., Winograd, J.M.,
Beheshti, M., Chavan, P.P., Criqui, M.H., Desai, R., Dhamminda Dharmaratne, S., 2008. Tumor formation following murine neural precursor cell transplantation in a
Dorsey, E.R., Wilder Eagan, A., Elgendy, I.Y., et al., 2021. Burden of neurological rat peripheral nerve injury model. J. Reconstr. Microsurg. 24 (8), 545–550.
disorders across the US from 1990-2017: a global burden of disease study. JAMA Kalladka, D., Sinden, J., Pollock, K., Haig, C., McLean, J., Smith, W., McConnachie, A.,
Neurol. 78 (2), 165–176. Santosh, C., Bath, P.M., Dunn, L., Muir, K.W., 2016. Human neural stem cells in
Feng, Z., Liu, J., Ju, R., 2013. Hyperbaric oxygen treatment promotes neural stem cell patients with chronic ischaemic stroke (PISCES): a phase 1, first-in-man study.
proliferation in the subventricular zone of neonatal rats with hypoxic-ischemic brain Lancet (London, England) 388 (10046), 787–796.
damage. Neural Regen. Res. 8 (13), 1220–1227. Kalluri, H.S.G., Dempsey, R.J., 2008. Growth factors, stem cells, and stroke. Neurosurg.
Ford, E., Pearlman, J., Ruan, T., Manion, J., Waller, M., Neely, G.G., Caron, L., 2020. Focus. 24 (3–4). E14.
Human Pluripotent Stem cells-based therapies for neurodegenerative diseases: Kanazawa, M., Miura, M., Toriyabe, M., Koyama, M., Hatakeyama, M., Ishikawa, M.,
current status and challenges. Cells 9 (11). Nakajima, T., Onodera, O., Takahashi, T., Nishizawa, M., Shimohata, T., 2017.
Freed, C.R., Greene, P.E., Breeze, R.E., Tsai, W.Y., DuMouchel, W., Kao, R., Dillon, S., Microglia preconditioned by oxygen-glucose deprivation promote functional
Winfield, H., Culver, S., Trojanowski, J.Q., Eidelberg, D., Fahn, S., 2001. recovery in ischemic rats. Sci. Rep. 7, 42582.
Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. Kang, J.M., Yeon, B.K., Cho, S.-J., Suh, Y.-H., 2016. Stem cell therapy for Alzheimer’s
N. Engl. J. Med. 344 (10), 710–719. disease: a review of recent clinical trials. J. Alzheimer’s Dis. JAD 54 (3), 879–889.
Gao, L., Peng, Y., Xu, W., He, P., Li, T., Lu, X., Chen, G., 2020. Progress in stem cell Kawabori, M., Shichinohe, H., Kuroda, S., Houkin, K., 2020. Clinical trials of stem cell
therapy for spinal cord injury. Stem Cells Int. 2020, 2853650. therapy for cerebral ischemic stroke. Int. J. Mol. Sci. 21 (19).
Gao, X., Han, Z., Huang, C., Lei, H., Li, G., Chen, L., Feng, D., Zhou, Z., Shi, Q., Cheng, L., Kim, H.-J., 2011. Stem cell potential in Parkinson’s disease and molecular factors for the
Zhou, X., 2022. An anti-inflammatory and neuroprotective biomimetic nanoplatform generation of dopamine neurons. Biochim. Biophys. Acta 1812 (1), 1–11.
for repairing spinal cord injury. Bioactive Mater. 18, 569–582. Kim, C., Lee, H.C., Sung, J.-J., 2014. Amyotrophic lateral sclerosis - cell based therapy
Gao, X., You, Z., Li, Y., Kang, X., Yang, W., Wang, H., Zhang, T., Zhao, X., Sun, Y., and novel therapeutic development. Exp. Neurobiol. 23 (3), 207–214.
Shen, H., Dai, J., 2023. Multifunctional hydrogel modulates the immune Kim, H.J., Seo, S.W., Chang, J.W., Lee, J.I., Kim, C.H., Chin, J., Choi, S.J., Kwon, H.,
microenvironment to improve allogeneic spinal cord tissue survival for complete Yun, H.J., Lee, J.M., Kim, S.T., Choe, Y.S., Lee, K.-H., Na, D.L., 2015. Stereotactic
spinal cord injury repair. Acta Biomater. 155, 235–246. brain injection of human umbilical cord blood mesenchymal stem cells in patients
Gaudet, A.D., Fonken, L.K., 2018. Glial cells shape pathology and repair after spinal cord with Alzheimer’s disease dementia: A phase 1 clinical trial. Alzheimer’s Dementia
injury. Neurotherapeutics: J. Am. Soc. Exp. NeuroTherap. 15 (3), 554–577. (New York, N. Y.) 1 (2), 95–102.
Ghaddar, B., Lübke, L., Couret, D., Rastegar, S., Diotel, N., 2021. Cellular mechanisms Kizil, C., Kyritsis, N., Brand, M., 2015. Effects of inflammation on stem cells: together
participating in brain repair of adult zebrafish and mammals after injury. Cells 10 they strive? EMBO Rep. 16 (4), 416–426.
(2). Koffler, J., Zhu, W., Qu, X., Platoshyn, O., Dulin, J.N., Brock, J., Graham, L., Lu, P.,
Giacomelli, E., Vahsen, B.F., Calder, E.L., Xu, Y., Scaber, J., Gray, E., Dafinca, R., Sakamoto, J., Marsala, M., Chen, S., Tuszynski, M.H., 2019. Biomimetic 3D-printed
Talbot, K., Studer, L., 2022. Human stem cell models of neurodegeneration: from scaffolds for spinal cord injury repair. Nat. Med. 25 (2), 263–269.
basic science of amyotrophic lateral sclerosis to clinical translation. Cell Stem Cell 29 Kyritsis, N., Kizil, C., Zocher, S., Kroehne, V., Kaslin, J., Freudenreich, D., Iltzsche, A.,
(1), 11–35. Brand, M., 2012. Acute inflammation initiates the regenerative response in the adult
Gilbert, E.A.B., Lakshman, N., Lau, K.S.K., Morshead, C.M., 2022. Regulating endogenous zebrafish brain. Science (New York, N.Y.) 338 (6112), 1353–1356.
neural stem cell activation to promote spinal cord injury repair. Cells 11 (5). Lee, H.-L., Lee, H.Y., Yun, Y., Oh, J., Che, L., Lee, M., Ha, Y., 2016. Hypoxia-specific,
Golas, M.M., Sander, B., 2016. Use of human stem cells in Huntington disease modeling VEGF-expressing neural stem cell therapy for safe and effective treatment of
and translational research. Exp. Neurol. 278, 76–90. neuropathic pain. J. Controll. Rel. Off. J. Controlled Release Soc. 226, 21–34.
Grochowski, C., Radzikowska, E., Maciejewski, R., 2018. Neural stem cell therapy-brief Levenberg, S., Burdick, J.A., Kraehenbuehl, T., Langer, R., 2005. Neurotrophin-induced
review. Clin. Neurol. Neurosurg. 173, 8–14. differentiation of human embryonic stem cells on three-dimensional polymeric
Guan, J., Zhu, Z., Zhao, R.C., Xiao, Z., Wu, C., Han, Q., Chen, L., Tong, W., Zhang, J., scaffolds. Tissue Eng. 11 (3–4), 506–512.
Han, Q., Gao, J., Feng, M., Bao, X., Dai, J., et al., 2013. Transplantation of human Li, G., Zhang, B., Sun, J.-H., Shi, L.-Y., Huang, M.-Y., Huang, L.-J., Lin, Z.-J., Lin, Q.-Y.,
mesenchymal stem cells loaded on collagen scaffolds for the treatment of traumatic Lai, B.-Q., Ma, Y.-H., Jiang, B., Ding, Y., Zhang, H.-B., Li, M.-X., et al., 2021. An NT-
brain injury in rats. Biomaterials 34 (24), 5937–5946. 3-releasing bioscaffold supports the formation of TrkC-modified neural stem cell-
Hankey, G.J., 2017. Stroke. Lancet (London, England) 389 (10069), 641–654. derived neural network tissue with efficacy in repairing spinal cord injury. Bioactive
Hapner, S.J., Nielsen, K.M., Chaverra, M., Esper, R.M., Loeb, J.A., Lefcort, F., 2006. NT-3 Mater. 6 (11), 3766–3781.
and CNTF exert dose-dependent, pleiotropic effects on cells in the immature dorsal Li, W., Mandeville, E.T., Durán-Laforet, V., Fukuda, N., Yu, Z., Zheng, Y., Held, A.,
root ganglion: neuregulin-mediated proliferation of progenitor cells and neuronal Park, J.-H., Nakano, T., Tanaka, M., Shi, J., Esposito, E., Niu, W., Xing, C., et al.,
differentiation. Dev. Biol. 297 (1), 182–197. 2022. Endothelial cells regulate astrocyte to neural progenitor cell trans-
Hargus, G., Cooper, O., Deleidi, M., Levy, A., Lee, K., Marlow, E., Yow, A., Soldner, F., differentiation in a mouse model of stroke. Nat. Commun. 13 (1), 7812.
Hockemeyer, D., Hallett, P.J., Osborn, T., Jaenisch, R., Isacson, O., 2010. Li, Y., Tran, A., Graham, L., Brock, J., Tuszynski, M.H., Lu, P., 2023. BDNF guides neural
Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the stem cell-derived axons to ventral interneurons and motor neurons after spinal cord
adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc. Natl. injury. Exp. Neurol. 359, 114259.
Acad. Sci. U. S. A. 107 (36), 15921–15926. Liau, L.L., Looi, Q.H., Chia, W.C., Subramaniam, T., Ng, M.H., Law, J.X., 2020. Treatment
Hennemann, B., Ickenstein, G., Sauerbruch, S., Luecke, K., Haas, S., Horn, M., of spinal cord injury with mesenchymal stem cells. Cell Biosci. 10, 112.
Andreesen, R., Bogdahn, U., Winkler, J., 2008. Mobilization of CD34+ Lilja, A.M., Malmsten, L., Röjdner, J., Voytenko, L., Verkhratsky, A., Ögren, S.O.,
hematopoietic cells, colony-forming cells and long-term culture-initiating cells into Nordberg, A., Marutle, A., 2015. Neural stem cell transplant-induced effect on
the peripheral blood of patients with an acute cerebral ischemic insult. Cytotherapy neurogenesis and cognition in Alzheimer Tg2576 mice is inhibited by concomitant
10 (3), 303–311. treatment with amyloid-lowering or cholinergic α7 nicotinic receptor drugs. Neural
Huang, L., Zhang, L., 2019. Neural stem cell therapies and hypoxic-ischemic brain injury. Plasticity 2015, 370432.
Prog. Neurobiol. 173, 1–17. Liu, X.-Y., Yang, L.-P., Zhao, L., 2020. Stem cell therapy for Alzheimer’s disease. World J.
Huang, D., Cao, Y., Yang, X., Liu, Y., Zhang, Y., Li, C., Chen, G., Wang, Q., 2021. Stem Cells 12 (8), 787–802.
A nanoformulation-mediated multifunctional stem cell therapy with improved beta- Liu, X., Song, S., Chen, Z., Gao, C., Li, Y., Luo, Y., Huang, J., Zhang, Z., 2022. Release of
amyloid clearance and neural regeneration for Alzheimer’s disease. Adv. Mater. O-GlcNAc transferase inhibitor promotes neuronal differentiation of neural stem
(Deerfield Beach, Fla.) 33 (13) e2006357. cells in 3D bioprinted supramolecular hydrogel scaffold for spinal cord injury repair.
Huang, Y., Ren, H., Gao, X., Cai, D., Shan, H., Bai, J., Sheng, L., Jin, Y., Zhou, X., 2022. Acta Biomater. 151, 148–162.
Amlodipine improves spinal cord injury repair by inhibiting motoneuronal apoptosis Longhi, L., Zanier, E.R., Royo, N., Stocchetti, N., McIntosh, T.K., 2005. Stem cell
through autophagy upregulation. Spine 47 (17), E570–E578. transplantation as a therapeutic strategy for traumatic brain injury. Transpl.
Huebner, E.A., Strittmatter, S.M., 2009. Axon regeneration in the peripheral and central Immunol. 15 (2), 143–148.
nervous systems. Results Probl. Cell Differ. 48, 339–351. Lu, P., Freria, C.M., Graham, L., Tran, A.N., Villarta, A., Yassin, D., Huie, J.R.,
Hulisz, D., 2018. Amyotrophic lateral sclerosis: disease state overview. Am. J. Manag. Ferguson, A.R., Tuszynski, M.H., 2022. Rehabilitation combined with neural
Care 24 (15 Suppl), S320–S326. progenitor cell grafts enables functional recovery in chronic spinal cord injury. JCI
Huo, W., Liu, X., Tan, C., Han, Y., Kang, C., Quan, W., Chen, J., 2014. Stem cell Insight 7 (16).
transplantation for treating stroke: status, trends and development. Neural Regen. Lugert, S., Basak, O., Knuckles, P., Haussler, U., Fabel, K., Götz, M., Haas, C.A.,
Res. 9 (17), 1643–1648. Kempermann, G., Taylor, V., Giachino, C., 2010. Quiescent and active hippocampal

10
C. Ying et al. Experimental Neurology 369 (2023) 114543

neural stem cells with distinct morphologies respond selectively to physiological and patients with amyotrophic lateral sclerosis. Front. Biosci. (Landmark edition) 26
pathological stimuli and aging. Cell Stem Cell 6 (5), 445–456. (10), 693–706.
Lukovic, D., Stojkovic, M., Moreno-Manzano, V., Jendelova, P., Sykova, E., Phanstiel, D.H., Brumbaugh, J., Wenger, C.D., Tian, S., Probasco, M.D., Bailey, D.J.,
Bhattacharya, S.S., Erceg, S., 2015. Concise review: reactive astrocytes and stem cells Swaney, D.L., Tervo, M.A., Bolin, J.M., Ruotti, V., Stewart, R., Thomson, J.A.,
in spinal cord injury: good guys or bad guys? Stem Cells (Dayton, Ohio) 33 (4), Coon, J.J., 2011. Proteomic and phosphoproteomic comparison of human ES and iPS
1036–1041. cells. Nat. Methods 8 (10), 821–827.
Lunn, J.S., Sakowski, S.A., Feldman, E.L., 2014. Concise review: stem cell therapies for Piccini, P., Brooks, D.J., Björklund, A., Gunn, R.N., Grasby, P.M., Rimoldi, O.,
amyotrophic lateral sclerosis: recent advances and prospects for the future. Stem Brundin, P., Hagell, P., Rehncrona, S., Widner, H., Lindvall, O., 1999. Dopamine
Cells (Dayton, Ohio) 32 (5), 1099–1109. release from nigral transplants visualized in vivo in a Parkinson’s patient. Nat.
Ma, K., Deng, X., Xia, X., Fan, Z., Qi, X., Wang, Y., Li, Y., Ma, Y., Chen, Q., Peng, H., Neurosci. 2 (12), 1137–1140.
Ding, J., Li, C., Huang, Y., Tian, C., et al., 2018. Direct conversion of mouse Ralli, M., Lambiase, A., Artico, M., de Vincentiis, M., Greco, A., 2019. Amyotrophic
astrocytes into neural progenitor cells and specific lineages of neurons. Translat. lateral sclerosis: autoimmune pathogenic mechanisms, clinical features, and
Neurodegenerat. 7, 29. therapeutic perspectives. Israel Med. Assoc. J. IMAJ 21 (7), 438–443.
Martínez-Morales, P.L., Revilla, A., Ocaña, I., González, C., Sainz, P., McGuire, D., Ratajczak, M.Z., Ratajczak, J., Kucia, M., 2019. Very small embryonic-like stem cells
Liste, I., 2013. Progress in stem cell therapy for major human neurological disorders. (VSELs). Circ. Res. 124 (2), 208–210.
Stem Cell Rev. Rep. 9 (5), 685–699. Reich, S.G., Savitt, J.M., 2019. Parkinson’s disease. Med. Clin. North Am. 103 (2),
Mazzini, L., Gelati, M., Profico, D.C., Sorarù, G., Ferrari, D., Copetti, M., Muzi, G., 337–350.
Ricciolini, C., Carletti, S., Giorgi, C., Spera, C., Frondizi, D., Masiero, S., Stecco, A., Ritfeld, G.J., Roos, R.A.C., Oudega, M., 2011. Stem cells for central nervous system repair
et al., 2019. Results from phase I clinical trial with Intraspinal injection of neural and rehabilitation. PM & R : J. Injury Funct. Rehabilit. 3 (6 Suppl 1), S117–S122.
stem cells in amyotrophic lateral sclerosis: a Long-term outcome. Stem Cells Transl. Rosenzweig, E.S., Brock, J.H., Lu, P., Kumamaru, H., Salegio, E.A., Kadoya, K., Weber, J.
Med. 8 (9), 887–897. L., Liang, J.J., Moseanko, R., Hawbecker, S., Huie, J.R., Havton, L.A., Nout-Lomas, Y.
Menorca, R., Fussell, T.S., Elfar, J.C., 2013. Nerve physiology: mechanisms of injury and S., Ferguson, A.R., et al., 2018. Restorative effects of human neural stem cell grafts
recovery. Hand Clin. 29 (3), 317–330. on the primate spinal cord. Nat. Med. 24 (4), 484–490.
Moreno-Jiménez, E.P., Flor-García, M., Terreros-Roncal, J., Rábano, A., Cafini, F., Pallas- Rossignol, J., Fink, K., Davis, K., Clerc, S., Crane, A., Matchynski, J., Lowrance, S.,
Bazarra, N., Ávila, J., Llorens-Martín, M., 2019. Adult hippocampal neurogenesis is Bombard, M., Dekorver, N., Lescaudron, L., Dunbar, G.L., 2014. Transplants of adult
abundant in neurologically healthy subjects and drops sharply in patients with mesenchymal and neural stem cells provide neuroprotection and behavioral sparing
Alzheimer’s disease. Nat. Med. 25 (4), 554–560. in a transgenic rat model of Huntington’s disease. Stem Cells (Dayton, Ohio) 32 (2),
Mothe, A.J., Tator, C.H., 2012. Advances in stem cell therapy for spinal cord injury. 500–509.
J. Clin. Invest. 122 (11), 3824–3834. Sackett, S.D., Brown, M.E., Tremmel, D.M., Ellis, T., Burlingham, W.J., Odorico, J.S.,
Mueller, F.J., McKercher, S.R., Imitola, J., Loring, J.F., Yip, S., Khoury, S.J., Snyder, E.Y., 2016. Modulation of human allogeneic and syngeneic pluripotent stem cells and
2005. At the interface of the immune system and the nervous system: how immunological implications for transplantation. Transplant. Rev. (Orlando, Fla.) 30
neuroinflammation modulates the fate of neural progenitors in vivo. Ernst Schering (2), 61–70.
research foundation workshop 53, 83–114. Saeki, S., Tokutake, K., Takasu, M., Kurimoto, S., Asami, Y., Onaka, K., Saeki, M.,
Müller, J., Ossig, C., Greiner, J.F.W., Hauser, S., Fauser, M., Widera, D., Kaltschmidt, C., Hirata, H., 2022. Functional reconstruction of denervated muscle by
Storch, A., Kaltschmidt, B., 2015. Intrastriatal transplantation of adult human neural xenotransplantation of neural cells from porcine to rat. Int. J. Mol. Sci. 23, 15.
crest-derived stem cells improves functional outcome in parkinsonian rats. Stem Sakai, K., Yamamoto, A., Matsubara, K., Nakamura, S., Naruse, M., Yamagata, M.,
Cells Transl. Med. 4 (1), 31–43. Sakamoto, K., Tauchi, R., Wakao, N., Imagama, S., Hibi, H., Kadomatsu, K.,
Musiał, A., Bajda, M., Malawska, B., 2007. Recent developments in cholinesterases Ishiguro, N., Ueda, M., 2012. Human dental pulp-derived stem cells promote
inhibitors for Alzheimer’s disease treatment. Curr. Med. Chem. 14 (25), 2654–2679. locomotor recovery after complete transection of the rat spinal cord by multiple
Nakagomi, T., Molnár, Z., Taguchi, A., Nakano-Doi, A., Lu, S., Kasahara, Y., neuro-regenerative mechanisms. J. Clin. Invest. 122 (1), 80–90.
Nakagomi, N., Matsuyama, T., 2012. Leptomeningeal-derived doublecortin- Sakowski, S.A., Chen, K.S., 2022. Stem cell therapy for central nervous system disorders:
expressing cells in poststroke brain. Stem Cells Dev. 21 (13), 2350–2354. metabolic interactions between transplanted cells and local microenvironments.
Nguyen, N., Lee, S.B., Lee, Y.S., Lee, K.-H., Ahn, J.-Y., 2009. Neuroprotection by NGF and Neurobiol. Dis. 173, 105842.
BDNF against neurotoxin-exerted apoptotic death in neural stem cells are mediated Sakthiswary, R., Raymond, A.A., 2012. Stem cell therapy in neurodegenerative diseases:
through Trk receptors, activating PI3-kinase and MAPK pathways. Neurochem. Res. from principles to practice. Neural Regen. Res. 7 (23), 1822–1831.
34 (5), 942–951. Scherbel, U., Raghupathi, R., Nakamura, M., Saatman, K.E., Trojanowski, J.Q.,
Nicole, O., Ali, C., Docagne, F., Plawinski, L., MacKenzie, E.T., Vivien, D., Buisson, A., Neugebauer, E., Marino, M.W., McIntosh, T.K., 1999. Differential acute and chronic
2001. Neuroprotection mediated by glial cell line-derived neurotrophic factor: responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc.
involvement of a reduction of NMDA-induced calcium influx by the mitogen- Natl. Acad. Sci. U. S. A. 96 (15), 8721–8726.
activated protein kinase pathway. J. Neurosci. Off. J. Soc. Neurosci. 21 (9), Shahbazi, E., Mirakhori, F., Ezzatizadeh, Baharvand, H., 2018. Reprogramming of
3024–3033. somatic cells to induced neural stem cells. Methods (San Diego, Calif.) 133, 21–28.
Nout-Lomas, Y.S., 2022. Traumatic nervous system injury. the veterinary clinics of North Shao, A., Tu, S., Lu, J., Zhang, J., 2019. Crosstalk between stem cell and spinal cord
America. Equine Pract. 38 (2), 363–377. injury: pathophysiology and treatment strategies. Stem Cell Res Ther 10 (1), 238.
Nowicka, N., Juranek, J., Juranek, J.K., Wojtkiewicz, J., 2019. Risk factors and emerging Shi, Y., Zhou, L., Tian, J., Wang, Y., 2009. Transplantation of neural stem cells
therapies in amyotrophic lateral sclerosis. Int. J. Mol. Sci. 20 (11). overexpressing glia-derived neurotrophic factor promotes facial nerve regeneration.
Ogawa, Y., Sawamoto, K., Miyata, T., Miyao, S., Watanabe, M., Nakamura, M., Acta Otolaryngol. 129 (8), 906–914.
Bregman, B.S., Koike, M., Uchiyama, Y., Toyama, Y., Okano, H., 2002. Sivandzade, F., Cucullo, L., 2021. Regenerative stem cell therapy for neurodegenerative
Transplantation of in vitro-expanded fetal neural progenitor cells results in diseases: an overview. Int. J. Mol. Sci. 22 (4).
neurogenesis and functional recovery after spinal cord contusion injury in adult rats. Song, M., Kim, Y.-J., Kim, Y.-H., Roh, J., Kim, E.-C., Lee, H.J., Kim, S.U., Yoon, B.-W.,
J. Neurosci. Res. 69 (6), 925–933. 2015. Long-term effects of magnetically targeted ferumoxide-labeled human neural
Oki, K., Tatarishvili, J., Wood, J., Koch, P., Wattananit, S., Mine, Y., Monni, E., stem cells in focal cerebral ischemia. Cell Transplant. 24 (2), 183–190.
Tornero, D., Ahlenius, H., Ladewig, J., Brüstle, O., Lindvall, O., Kokaia, Z., 2012. Sonntag, K.-C., Song, B., Lee, N., Jung, J.H., Cha, Y., Leblanc, P., Neff, C., Kong, S.W.,
Human-induced pluripotent stem cells form functional neurons and improve Carter, B.S., Schweitzer, J., Kim, K.-S., 2018. Pluripotent stem cell-based therapy for
recovery after grafting in stroke-damaged brain. Stem Cells (Dayton, Ohio) 30 (6), Parkinson’s disease: current status and future prospects. Prog. Neurobiol. 168, 1–20.
1120–1133. Spalding, K.L., Bergmann, O., Alkass, K., Bernard, S., Salehpour, M., Huttner, H.B.,
Olanow, C.W., Goetz, C.G., Kordower, J.H., Stoessl, A.J., Sossi, V., Brin, M.F., Boström, E., Westerlund, I., Vial, C., Buchholz, B.A., Possnert, G., Mash, D.C.,
Shannon, K.M., Nauert, G.M., Perl, D.P., Godbold, J., Freeman, T.B., 2003. A double- Druid, H., Frisén, J., 2013. Dynamics of hippocampal neurogenesis in adult humans.
blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Cell 153 (6), 1219–1227.
Ann. Neurol. 54 (3), 403–414. Suda, S., Nito, C., Yokobori, S., Sakamoto, Y., Nakajima, M., Sowa, K., Obinata, H.,
Oliveira, S.L.B., Pillat, M.M., Cheffer, A., Lameu, C., Schwindt, T.T., Ulrich, H., 2013. Sasaki, K., Savitz, S.I., Kimura, K., 2020. Recent advances in cell-based therapies for
Functions of neurotrophins and growth factors in neurogenesis and brain repair. ischemic stroke. Int. J. Mol. Sci. 21 (18).
Cytometry. Part A J. Int. Soc. Analyt. Cytol. 83 (1), 76–89. Sudhakar, V., Richardson, R.M., 2019. Gene therapy for neurodegenerative diseases.
Oskarsson, B., Gendron, T.F., Staff, N.P, 2018. Amyotrophic lateral sclerosis: an update Neurotherapeutics: J. Am. Soc. Exp. NeuroTherap. 16 (1), 166–175.
for 2018. Mayo Clin. Proc. 93 (11), 1617–1628. Sugai, K., Sumida, M., Shofuda, T., Yamaguchi, R., Tamura, T., Kohzuki, T., Abe, T.,
Park, K.J., Park, E., Liu, E., Baker, A.J., 2014. Bone marrow-derived endothelial Shibata, R., Kamata, Y., Ito, S., Okubo, T., Tsuji, O., Nori, S., Nagoshi, N., et al.,
progenitor cells protect postischemic axons after traumatic brain injury. J. Cerebral 2021. First-in-human clinical trial of transplantation of iPSC-derived NS/PCs in
Blood Flow Metabol. Off. J. Int. Soc. Cerebral Blood Flow Metabol. 34 (2), 357–366. subacute complete spinal cord injury: study protocol. Regenerat. Therapy 18,
Park, D., Choi, E.-K., Cho, T.-H., Joo, S.S., Kim, Y.-B., 2020. Human neural stem cells 321–333.
encoding ChAT gene restore cognitive function via acetylcholine synthesis, Aβ Sugaya, K., Vaidya, M., 2018. Stem cell therapies for neurodegenerative diseases. Adv.
elimination, and neuroregeneration in APPswe/PS1dE9 mice. Int. J. Mol. Sci. 21 Exp. Med. Biol. 1056, 61–84.
(11). Sun, X., Zhang, C., Xu, J., Zhai, H., Liu, S., Xu, Y., Hu, Y., Long, H., Bai, Y., Quan, D.,
Perez-Asensio, F.J., Perpiñá, U., Planas, A.M., Pozas, E., 2013. Interleukin-10 regulates 2020. Neurotrophin-3-loaded multichannel nanofibrous scaffolds promoted anti-
progenitor differentiation and modulates neurogenesis in adult brain. J. Cell Sci. 126 inflammation, neuronal differentiation, and functional recovery after spinal cord
(Pt 18), 4208–4219. injury. ACS Biomater Sci. Eng. 6 (2), 1228–1238.
Petrou, P., Kassis, I., Yaghmour, N.E., Ginzberg, A., Karussis, D., 2021. A phase II clinical Takahashi, K., Yamanaka, S., 2006. Induction of pluripotent stem cells from mouse
trial with repeated intrathecal injections of autologous mesenchymal stem cells in embryonic and adult fibroblast cultures by defined factors. Cell 126 (4), 663–676.

11
C. Ying et al. Experimental Neurology 369 (2023) 114543

Tator, C.H., 2006. Review of treatment trials in human spinal cord injury: issues, Wang, Z., Luo, Y., Chen, L., Liang, W., 2017. Safety of neural stem cell transplantation in
difficulties, and recommendations. Neurosurgery 59 (5), 957–982 (discussion 982- patients with severe traumatic brain injury. Exp. Ther. Med. 13 (6), 3613–3618.
987). Wu, D., Zhang, Y., Xu, X., Guo, T., Xie, D., Zhu, R., Chen, S., Ramakrishna, S., He, L.,
Tazaki, A., Tanaka, E.M., Fei, J.-F., 2017. Salamander spinal cord regeneration: the 2018. RGD/TAT-functionalized chitosan-graft-PEI-PEG gene nanovector for
ultimate positive control in vertebrate spinal cord regeneration. Dev. Biol. 432 (1), sustained delivery of NT-3 for potential application in neural regeneration. Acta
63–71. Biomater. 72, 266–277.
Terry, R.D., Davies, P., 1980. Dementia of the Alzheimer type. Annu. Rev. Neurosci. 3, Xian, P., Hei, Y., Wang, R., Wang, T., Yang, J., Li, J., Di, Z., Liu, Z., Baskys, A., Liu, W.,
77–95. Wu, S., Long, Q., 2019. Mesenchymal stem cell-derived exosomes as a
Toda, T., Gage, F.H., 2018. Review: adult neurogenesis contributes to hippocampal nanotherapeutic agent for amelioration of inflammation-induced astrocyte
plasticity. Cell Tissue Res. 373 (3), 693–709. alterations in mice. Theranostics 9 (20), 5956–5975.
Tornero, D., Wattananit, S., Grønning Madsen, M., Koch, P., Wood, J., Tatarishvili, J., Xiong, Y., Mahmood, A., Chopp, M., 2009. Emerging treatments for traumatic brain
Mine, Y., Ge, R., Monni, E., Devaraju, K., Hevner, R.F., Brüstle, O., Lindvall, O., injury. Expert Opin. Emerg. Drugs 14 (1), 67–84.
Kokaia, Z., 2013. Human induced pluripotent stem cell-derived cortical neurons Yan, Z.-J., Hu, Y.-Q., Zhang, H.-T., Zhang, P., Xiao, Z.-Y., Sun, X.-L., Cai, Y.-Q., Hu, C.-C.,
integrate in stroke-injured cortex and improve functional recovery. Brain J. Neurol. Xu, R.-X., 2013. Comparison of the neural differentiation potential of human
136 (Pt 12), 3561–3577. mesenchymal stem cells from amniotic fluid and adult bone marrow. Cell. Mol.
Trounson, A., McDonald, C., 2015. Stem cell therapies in clinical trials: progress and Neurobiol. 33 (4), 465–475.
challenges. Cell Stem Cell 17 (1), 11–22. Yang, X., Ji, Y., Wang, W., Zhang, L., Chen, Z., Yu, M., Shen, Y., Ding, F., Gu, X., Sun, H.,
Uccelli, A., Pistoia, V., Moretta, L., 2007. Mesenchymal stem cells: a new strategy for 2021. Amyotrophic lateral sclerosis: molecular mechanisms, biomarkers, and
immunosuppression? Trends Immunol. 28 (5), 219–226. therapeutic strategies. Antioxidants (Basel, Switzerland) 10 (7).
Uchida, H., Morita, T., Niizuma, K., Kushida, Y., Kuroda, Y., Wakao, S., Sakata, H., Yang, S.-G., Wang, X.-W., Qian, C., Zhou, F.-Q., 2022. Reprogramming neurons for
Matsuzaka, Y., Mushiake, H., Tominaga, T., Borlongan, C.V., Dezawa, M., 2016. regeneration: the fountain of youth. Prog. Neurobiol. 214, 102284.
Transplantation of unique subpopulation of fibroblasts, muse cells, ameliorates Yu, D., Ma, M., Liu, Z., Pi, Z., Du, X., Ren, J., Qu, X., 2020. MOF-encapsulated nanozyme
experimental stroke possibly via robust neuronal differentiation. Stem Cells (Dayton, enhanced siRNA combo: control neural stem cell differentiation and ameliorate
Ohio) 34 (1), 160–173. cognitive impairments in Alzheimer’s disease model. Biomaterials 255, 120160.
Urbán, N., Blomfield, I.M., Guillemot, F., 2019. Quiescence of adult mammalian neural Yuan, J., Yankner, B.A., 2000. Apoptosis in the nervous system. Nature 407 (6805),
stem cells: a highly regulated rest. Neuron 104 (5), 834–848. 802–809.
Ustyantseva, E.I., Medvedev, S.P., Zakian, S.M., 2020. Studying ALS: current approaches, Zhang, M., Liu, Y., Shi, L., Fang, L., Xu, L., Cao, Y., 2022. Neural stemness unifies cell
effect on potential treatment strategy. Adv. Exp. Med. Biol. 1241, 195–217. tumorigenicity and pluripotent differentiation potential. J. Biol. Chem. 298 (7),
Uyeda, A., Muramatsu, R., 2020. Molecular mechanisms of central nervous system 102106.
axonal regeneration and remyelination: a review. Int. J. Mol. Sci. 21 (21). Zhao, Y., Xiao, Z., Chen, B., Dai, J., 2017a. The neuronal differentiation
Vitrac, A., Cloëz-Tayarani, I., 2018. Induced pluripotent stem cells as a tool to study microenvironment is essential for spinal cord injury repair. Organogenesis 13 (3),
brain circuits in autism-related disorders. Stem Cell Res Ther 9 (1), 226. 63–70.
Wakabayashi, K., Nagai, A., Sheikh, A.M., Shiota, Y., Narantuya, D., Watanabe, T., Zhao, Y.-Z., Jiang, X., Lin, Q., Xu, H.-L., Huang, Y.-D., Lu, C.-T., Cai, J., 2017b.
Masuda, J., Kobayashi, S., Kim, S.U., Yamaguchi, S., 2010. Transplantation of human Thermosensitive heparin-poloxamer hydrogels enhance the effects of GDNF on
mesenchymal stem cells promotes functional improvement and increased expression neuronal circuit remodeling and neuroprotection after spinal cord injury. J. Biomed.
of neurotrophic factors in a rat focal cerebral ischemia model. J. Neurosci. Res. 88 Mater. Res. A 105 (10), 2816–2829.
(5), 1017–1025. Zhao, T., Zhu, T., Xie, L., Li, Y., Xie, R., Xu, F., Tang, H., Zhu, J., 2022. Neural stem cells
Walshe, J.M., 2014. Central nervous system regeneration. Qjm Monthly J. Assoc. therapy for ischemic stroke: progress and challenges. Transl. Stroke Res. 13 (5),
Physicians 107 (8), 3–15. 665–675.
Wang, X., Fu, S., Wang, Y., Yu, P., Hu, J., Gu, W., Xu, X.-M., Lu, P., 2007. Interleukin- Zheng, Y., Mao, Y.-R., Yuan, T.-F., Xu, D.-S., Cheng, L.-M., 2020. Multimodal treatment
1beta mediates proliferation and differentiation of multipotent neural precursor cells for spinal cord injury: a sword of neuroregeneration upon neuromodulation. Neural
through the activation of SAPK/JNK pathway. Mol. Cell. Neurosci. 36 (3), 343–354. Regen. Res. 15 (8), 1437–1450.
Wang, B., Gao, Y., Xiao, Z., Chen, B., Han, J., Zhang, J., Wang, X., Dai, J., 2009. Erk1/2 Zhong, D., Cao, Y., Li, C.-J., Li, M., Rong, Z.-J., Jiang, L., Guo, Z., Lu, H.-B., Hu, J.-Z.,
promotes proliferation and inhibits neuronal differentiation of neural stem cells. 2020. Neural stem cell-derived exosomes facilitate spinal cord functional recovery
Neurosci. Lett. 461 (3), 252–257. after injury by promoting angiogenesis. Exp. Biol. Med. (Maywood, N.J.) 245 (1),
Wang, E., Gao, J., Yang, Q., Parsley, M.O., Dunn, T.J., Zhang, L., DeWitt, D.S., Denner, L., 54–65.
Prough, D.S., Wu, P., 2012. Molecular mechanisms underlying effects of neural stem Zhou, G., Wang, Y., Gao, S., Fu, X., Cao, Y., Peng, Y., Zhuang, J., Hu, J., Shao, A.,
cells against traumatic axonal injury. J. Neurotrauma 29 (2), 295–312. Wang, L., 2021. Potential mechanisms and perspectives in ischemic stroke treatment
using stem cell therapies. Front. Cell Develop. Biol. 9, 646927.

12

You might also like