Next Article in Journal
Comprehensive Analysis of CPA4 as a Poor Prognostic Biomarker Correlated with Immune Cells Infiltration in Bladder Cancer
Previous Article in Journal
Response of Annual Herbaceous Plant Leaching and Decomposition to Periodic Submergence in Mega-Reservoirs: Changes in Litter Nutrients and Soil Properties for Restoration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Therapy for Neurological Disorders: The Perspective of Promising Cells

School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
*
Author to whom correspondence should be addressed.
Biology 2021, 10(11), 1142; https://doi.org/10.3390/biology10111142
Submission received: 6 October 2021 / Revised: 5 November 2021 / Accepted: 5 November 2021 / Published: 6 November 2021
(This article belongs to the Section Neuroscience)

Abstract

:

Simple Summary

Cell therapy has become a powerful method for regenerative medicine. However, there has not been an ideal cell type and cell source for the treatment of neurological diseases such as Parkinson’s disease and Alzheimer’s disease. This review aims to introduce the potentials of different cells for treating neurological disorders by collecting the results from related clinical trials and recent animal studies. It is an overview of some promising cells that may be clinically used for neurological disorders. The characteristics of each cell type and the main mechanism of function are also described.

Abstract

Neurological disorders are big public health challenges that are afflicting hundreds of millions of people around the world. Although many conventional pharmacological therapies have been tested in patients, their therapeutic efficacies to alleviate their symptoms and slow down the course of the diseases are usually limited. Cell therapy has attracted the interest of many researchers in the last several decades and has brought new hope for treating neurological disorders. Moreover, numerous studies have shown promising results. However, none of the studies has led to a promising therapy for patients with neurological disorders, despite the ongoing and completed clinical trials. There are many factors that may affect the outcome of cell therapy for neurological disorders due to the complexity of the nervous system, especially cell types for transplantation and the specific disease for treatment. This paper provides a review of the various cell types from humans that may be clinically used for neurological disorders, based on their characteristics and current progress in related studies.

Graphical Abstract

1. Introduction

Neurological disorders are defined as diseases of the nervous system. There are more than 600 neurological disorders that can be divided into four categories: (1) sudden onset conditions, such as stroke; (2) intermittent conditions, such as epilepsy; (3) progressive conditions, such as Parkinson’s disease (PD); (4) stable neurological conditions, such as cerebral palsy. These diseases have become a big public health challenge and present a tremendous burden to individuals, their families, and society. Some of the disorders can be common and severe, including stroke, Huntington’s disease (HD), traumatic brain injury (TBI), spinal cord injury (SCI), epilepsy, PD, Alzheimer’s disease (AD), Lyme disease, cerebral palsy, ataxia, amyotrophic lateral sclerosis (ALS), hypoxic-ischemic encephalopathy (HIE) and multiple sclerosis (MS). Currently, although there are many conventional therapies that can alleviate their symptoms, such as levodopa or deep brain stimulation for PD [1], cholinesterase inhibitors, or memantine for AD [2], these treatments have failed to slow or reverse the progression of the diseases, in other words, these diseases are not curable by the conventional therapies.
A common feature of many severe neurological disorders is the loss and/or dysfunction of massive neural cells, especially neurons. Therefore, regenerative medicine, especially cell therapy, has become an intriguing field for researchers in the last several decades. Cell therapy is thought to be an excellent therapy for many neurological disorders acting by replacing dead cells or/and releasing protective factors to the damaged cells or/and modulating the lesion’s microenvironment in the nervous system [3,4,5]. With the rapidly expanding studies in this research area, cell therapy has shown its unique potentials in treating neurological disorders. Currently, various types of cells have been used in cell therapy studies, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), neural stem/progenitor cells (NSPCs), mesenchymal stromal/stem cells (MSCs), and olfactory ensheathing cells (OECs), etc. However, despite the promising results from numerous animal-based studies, the outcomes from clinical trials are normally not as significant as from animal studies. This may be due to the difference between animal models and the real disease of patients and the differences in nervous systems between animals and humans.
The outcome of cell therapy can be affected by many factors, including cell source, cell type, route of administration, and the target disease. The aim of this review is to summarize the cells that may be clinically used for neurological disorders, especially for the diseases mentioned above, and their characteristics and current progresses in related studies.

2. Cell Source: Autologous or Allogeneic?

The transplantable cells for patients can be divided into two sources: autologous and allogeneic. Autologous cells are usually more favorable than allogeneic cells in regenerative medicine as they avoid several issues that allogeneic cells may have, such as immune rejection or the use of immunosuppressants, ethical issues, and finding suitable donors [6]. However, allogeneic transplants are necessary for the cells that are therapeutic but hard to obtain from the patients themselves, such as ESCs [7], NSPCs [8], and fetal stem cells (FSCs) that contain various stem cells from fetal blood and tissues (e.g., MSCs, NSPCs, and hematopoietic stem cells) (see Table 1). Nowadays, cell reprogramming provides a new opportunity to generate any type of autologous cells from another type of easily obtainable somatic cells, such as iPSCs from urine [9] and induced neural stem cells from fibroblasts [10]. However, the safety of these cells is highly related to the methods and vectors used in induction protocols [11,12,13], and the safety and therapeutic effects need to be well studied before taking into clinical application.
Interestingly, some cells, such as umbilical cord blood mononuclear cells and adipose-derived mesenchymal stem cells (hADSCs), have been reported as immature immune cells; namely, they will not cause immune rejection, even in the absence of immune suppression [14,15], which may make them suitable candidates as allogeneic transplants.

3. Different Cell Types and the Current Progress

3.1. Embryonic Stem Cells (hESCs)

Human ESCs (hESCs) are pluripotent cells that are derived from human embryos and are capable of self-renewing and differentiating into all types of human cells. The first cell line of hESCs was isolated by James Thomson in 1998 [16], which then brought a new hope of developing new hESC lines for cell therapy. The therapeutic potential of ESCs for neurological disorders was proved a long time ago [17,18]. However, several critical issues of hESCs have limited the related studies and clinical applications: (1) ethical issues, as the establishment of hESCs lines involves the exploitation and destruction of human embryos; (2) immune rejection. As hESCs are allogeneic cells, they may cause immune rejection after transplantation [19], or the patient may need a life-long administration of immunosuppressants; (3) tumor formation. Due to the pluripotency of hESCs, they have the potential to generate teratoma after transplantation [20,21]. Several methods have been reported to reduce or eliminate the tumorigenicity of ESCs, such as pre-treating ESCs with mitomycin [22], co-transplantation ESCs with MSCs [23]. Among these methods, pre-differentiating ESCs into target cells prior to transplantation is a favorable way. hESCs are normally taken as a source of different cells in vitro. hESCs derivates, such as hESCs-derived oligodendrocytes, hESCs-derived dopaminergic neurons, have shown their therapeutic effects in animal studies for treating neurological disorders [24,25]. Although several animal studies transplanting hESCs for neurological disorders (such as SCI [26]) have shown some therapeutic effects, hESCs are normally considered unlikely to be directly grafted for cell therapy. Remarkably, however, Geeta Shroff has transplanted hESCs via a similar cell delivery method into patients with Lyme disease, multiple sclerosis, spinal cord injury, stroke, or cerebral palsy, and has proved the effectiveness and safety of the cells (see Table 2), with some ethical concerns [27,28]. Moreover, it has been claimed that these hESCs prepared at their institute are unlikely to cause immune rejection even without immunosuppressants as they were harvested at the very initial stage of blastocyst when the genesis was not activated [7]. To summarize, hESCs are more used as a source of other transplantable cells rather than a direct transplantation candidate in cell therapy studies. However, a certain line of hESCs may become a suitable candidate for cell therapy for neurological disorders if the critical issues can be overcome in the future.

3.2. Induced Pluripotent Stem Cells (iPSCs)

The first human iPSCs (hiPSCs) line was generated by Takahashi et al. [35] after they introduced four factors (Oct3/4, Sox2, Klf4, and c-Myc) into human fibroblasts. hiPSCs share similarities with hESCs in many aspects such as proliferation, morphology, gene expression, and differentiation. This iPSCs technique (also called cell reprogramming) provides an opportunity to generate any type of patient-specific cells from other obtainable cells, such as fibroblasts or urine cells. Therefore, there are no ethical issues from hiPSCs. Moreover, autologous cells transplantation from patients themselves has no risk of immune rejection and no need to use immunosuppressants. However, there is still the risk of tumor formation [21]. iPSCs are classically generated by the integration of transcription factors with a viral vector, which may cause tumorigenesis and/or unpredictable mutagenesis in the genome. hiPSCs generated by non-integration methods, such as mRNA [36], plasmid [37], small molecules [38], are safer candidates for transplantation, although they are normally with comparatively low reprogramming efficiency [39].
To date, although several animal studies grafting hiPSCs have shown their therapeutic effects for neurological disorders, such as stroke and SCI [5,40] (see Table S1), similar to hESCs, hiPSCs are also usually taken as a source of other transplantable cells in vitro. Pre-differentiated hiPSCs, including hiPSCs-derived NSPCs, hiPSCs-derived neurons and hiPSCs-derived MSCs have been shown effective for neurological disorders [41,42,43]. Especially, several clinical trials based on hiPSCs derivates have been launched for the treatment of PD and SCI [44,45], and thus have deeply encouraged the use of hiPSCs in treating neurological disorders. However, it is noteworthy that hiPSCs-derived NSPCs may still have the potential to generate tumors [46]. The safety issues, including tumorigenicity and aberrant reprogramming of the cells, need to be clearly addressed before their clinical application.

3.3. Neural Stem/Progenitor Cells (NSPCs)

NSPCs are multipotent cells that can self-renew and generate the main types of cells making up the central nervous system (CNS), including neurons, oligodendrocytes, and astrocytes. NSPCs have a lower risk of tumor formation compared to ESCs and iPSCs as they are more specialized cells and have less self-renewing ability. Therefore, human NSPCs (hNSPCs) are considered a favorable candidate for treating neurological disorders. Although it has been shown that a subset of NSPCs is present in highly restricted regions during adult life, their proliferation declines with aging [47,48], and it is nearly impossible to isolate autologous hNSPCs for cell therapy. The common sources of hNSPCs for research are human fetuses, hESCs, hiPSCs, or hMSCs. Like hESCs, hNSPCs directly isolated from embryos or differentiated from hESCs also have ethical concerns and may cause immune rejection, and hNSPCs from hiPSCs may have the same issues as hiPSCs as mentioned above. hNSPCs can also be obtained by trans-differentiation from hMSCs under specific experimental conditions [49]. In addition, hMSCs-derived hNSPCs are normally preferred in clinical studies as they normally have fewer issues than those from hESCs or hiPSCs. However, it is still important to generate a stable, efficient, and standardized protocol to convert hMSCs to hNSPCs and study the similarity between hMSCs-derived hNSPCs and bona fide hNSPCs.
Currently, hundreds of animal studies have used hNSPCs for treating various neurological disorders, including stroke, SCI, TBI, PD, HD (see Table S2), which are far more than hESCs or hiPSCs transplantation studies. Moreover, their effectiveness has been proved and reported better than human MSCs in some conditions [50,51]. The grafted hNSPCs can not only differentiate into neurons and glia cells and establish a graft-host connection [52,53] but also produce trophic factors and modulate lesion microenvironment to improve behavior recovery [54,55]. Intriguingly, even the injection of hNSPCs secretome alone has been reported to support the functional recovery of 6-hydroxydopamine (6-OHDA) PD rats [56]. Many methods have been applied to enhance the therapeutic effects of hNSPCs, such as genetically modified hNSPCs by overexpressing a selected trophic factor. Brain-derived neurotrophic factor (BDNF)-overexpressing hNSPCs, glial cell line-derived neurotrophic factor (GDNF)-overexpressing hNSPCs, and insulin-like growth factor 1 (IGF-1)-overexpressing hNSPCs have all been shown significant therapeutic effects on neurological disorders [57,58,59]. In addition, pre-treating hNSPCs with a gamma-secretase inhibitor [60], metformin [61] or tumor necrosis factor α (TNFα) [62], co-transplantation of hNSPCs with MSCs [63] or using biomaterial scaffolds as a carrier for hNSPCs [64,65] have also been reported to improve their therapeutic potential for neurological diseases. However, it is worth noting that the subtype of hNSPCs may also influence their therapeutic effects for a specific disease. For example, the human fetal spinal cord-derived NSPCs or spinal cord-type NSPCs from hiPSCs have been shown to improve motor functions after SCI, but not human fetal brain-derived NSPCs or forebrain-type NSPCs from hiPSCs [64,66].
To date, some clinical studies have been accomplished, and a few have shown beneficial outcomes after hNSPCs transplantation for neurological disorders (see Table 3). For allogeneic transplantation, focusing on specific hNSPC lines may help to keep the consistency of the outcomes from bench to beside. Several clinical-grade hNSPC lines have been applied in clinical trials, including NSI-566. NSI-566 cell line has been shown to be safe and potential effective in ALS patients [67,68] and now is in a phase 3 trial for treating ALS. Moreover, the NSI-566 cell line has also been reported to significantly improve the behavioral and histological recovery of ischemic stroke patients [69]. On the other hand, hNSPCs from autologous MSCs are a preferred candidate for clinical therapies of neurological disorders and have been applied for treating cerebral palsy, MS and TBI, in clinical trials [70,71,72,73]. However, most of the trials are in phase 1 or 2 stages, which have proved the safety but not the statistical therapeutic effects of the cells. Further studies need to be performed to better understand the therapeutic effects of the cells for neurological disorders.
Induced neural stem/progenitor cells (iNSPCs), such as iPSCs, are generated from other types of somatic cells (such as fibroblasts or urine cells) but bypass the pluripotency stage. Generation of hiNSPCs is an attractive field and is more favorable than hiPSCs in treating neural diseases, as they are easier to differentiate into terminal neural cells and less tumorigenic. Currently, many hiNSPC lines have been generated through different methods [74,75], and several lines have been tested in animal disease models, such as SCI, glioblastoma, and stoke and have shown their significant therapeutic effects [76,77,78,79]. However, the safety and therapeutic potential of hiNSPC are highly related to the induction protocol and need to be further studied. Therefore, it is essential to establish a safe, stable, and efficient protocol to generate hiNSPC for cell therapy studies.

3.4. Neurons, Oligodendrocytes, and Astrocytes

Neurons, oligodendrocytes, and astrocytes are three main cell types that form the CNS and originate from a common lineage of hNSPCs during development. These cells are terminal cells from NSPCs differentiation and can also be obtained from human embryos, hESCs, hiPSCs, or hMSCs in cell therapy studies. Transplantation of these pre-differentiated cells can avoid unexpected or unwanted differentiation or tumor formation of stem cells in vivo, especially in neurological disorders that are associated with the loss or dysfunction of specific types of neural cells, such as transplantation of dopaminergic neurons or dopaminergic progenitors for PD. It has been reported that pre-differentiated GABAergic neurons from hNSPCs have shown greater repopulation of the damaged brain and better neurogenic activity and functional recovery than hNSPCs in a stroke model after transplantation, while hNSPCs have predominantly differentiated into astrocytes [86].
Because neurons are the main functional cells in the nervous system, they are usually more attractive than glia cells for regenerative medicine in neuroscience research. Studies have transplanted neurons or neuron progenitors into different disease models and have shown their therapeutic outcomes (see Table S3). However, it is believed that more mature/differentiated donor cells have less survival capacity after transplantation [87,88,89]. Therefore, for a specific neuron type transplantation (for example, dopaminergic neurons), an immature stage of the cells may lead to better outcomes than mature cells or progenitors [89,90,91]. In addition, as the pre-differentiated cells have less proliferation capacity, they may need more cells for transplantation to reach a therapeutic level. Remarkably, several clinical trials transplanting neurons or neuron-contained tissue have proved the survival and potential therapeutic effects in patients (see Table 4). Moreover, direct reprogramming of human neurons from other somatic cells may also provide a suitable source of therapeutic neurons for cell therapies [92,93].
Oligodendrocytes and astrocytes are glia cells that are thought of as supportive and protective cells in the nervous system. A recent study has shown that the conditioned medium from hiPSCs-derived glial progenitor cells could result in better therapeutic effects than that from hiPSCs-derived neuronal progenitor cells and show a higher content of neurotrophins, indicating their potentials for cell therapy [94]. The main function of oligodendrocytes is to form myelin sheaths to support and insulate axons. Therefore, oligodendrocytes are highly related to diseases involving demyelination, such as MS and ALS. It has been suggested that transplantation of human oligodendrocyte precursor cells (hOPCs) could result in notable therapeutic outcomes in animals with MS or SCI [95,96,97], whereas transplantation of mature oligodendrocytes has failed to remyelinate naked axons in SCI [98,99]. Therefore, transplantation of hOPCs may be a potential therapy for demyelinating diseases. On the other hand, astrocytes are the most numerous cell type in the brain, playing various functions, including maintaining homeostasis, providing neurotrophic support, and connecting neurons with the bloodstream [100]. The loss or dysfunction of astrocytes is related to many neurological disorders, such as stroke, epilepsy, and MS [101]. Moreover, the astrocyte-formed glial scars are a reason that prevents neuroregeneration after CNS injury [102]. However, a study has also reported functional recovery after grafting hiPSCs-derived astrocytes with overexpressing the major glutamate transporter, GLT1, in an SCI model, suggesting their pro-regenerative function [103]. Notably, it is reported that the therapeutic effects of astrocytes are highly related to the subtypes of the cells. Stephen et al. found that astrocytes generated from human glial precursor cells by exposure to bone morphogenetic protein could promote significant functional recovery after SCI, whereas astrocytes generated by exposing the cells to ciliary neurotrophic factor failed to generate similar results [104]. Overall, the number of transplantation studies targeting oligodendrocytes or astrocytes is much lower than neuron-based studies. However, with better understanding of their functions in CNS, and more accessible sources of human oligodendrocytes or astrocytes (e.g., from hiPSCs), transplantation of oligodendrocytes or astrocytes may be a promising approach for treating specific neurological disorders.

3.5. Mesenchymal Stromal/Stem Cells (MSCs)

MSCs are multipotent cells that can differentiate into various cell types, including bone cells, muscle cells. In addition, it has been reported that MSCs can be transdifferentiated into neural cells under specific conditions, indicating the potential of MSCs for treating neurological diseases [49,108]. Human MSCs (hMSCs) can be easily obtained from many sources, among which the most studied hMSCs are those from bone marrow, umbilical cord, and adipose tissue. Although the detailed differences in their biological characteristics from different sources, the easy acquisition and the common properties, such as the capacity of self-renewing, multi-lineage differentiation, low tumorigenicity, low immunogenicity, and immunoregulatory function, have made hMSCs a promising candidate for cell therapy. Moreover, as mentioned above, hMSCs can also be a source of transplantable neural cells in vitro in models of neurological disorders.
Due to these advantages, hMSCs are currently one of the favorite cell types in cell therapy studies involving treating neurological diseases. Hundreds of animal studies have proved their safety and therapeutic effects for various neurological disorders (see Table S4). Moreover, even hMSC-conditioned medium or hMSCs-derived exosomes have been shown to alleviate the symptoms of experimental stroke or SCI [109,110]. Notably, although it has been reported that hMSCs can differentiate into neural cells in the injured area after transplantation [111,112], other researchers have also found that hMSCs could result in significant functional recovery without neural differentiation or even when hMSC is cleared away at the end of the experiment, suggesting that the main mechanisms of their beneficial functions are neurogenesis and angiogenesis promotion, anti-apoptosis, anti-inflammation, and immunomodulation rather than cell replacement [113,114,115,116,117]. Furthermore, studies have demonstrated that hMSCs can cross the blood-brain barrier (BBB) and home to the injured site through intravenous administration, which is a more suitable route for clinical use compared to direct local delivery to the affected tissue [118,119]. However, it is noteworthy that negative outcomes from hMSCs transplantation have also been shown in several studies, suggesting that administration of hMSCs alone may not be enough to generate significant therapeutic outcomes in some cases [120,121]. Therefore, several methods have been used to improve their therapeutic functions, such as pre-treatment [122], genetic modification [123], co-transplantation with hNSPCs [63], combined with other treatments [124], and using biomaterial scaffolds as a carrier [125]. Interestingly, the therapeutic effects of hMSCs may also be related to the source of the cells or the age of the donor. Jumpei et al. have reported that human cranial bone-derived MSCs could result in significant functional recovery in a rat model of stroke, but not human iliac bone-derived MSCs [126]. Susumu et al. have reported that transplantation of hMSCs from young donors could provide better functional recovery through multiple mechanisms than old hMSCs [127].
To date, many clinical trials using umbilical cord-derived MSCs (UC-MSCs) or autologous bone marrow (BM-MSCs) or adipose-derived MSCs (hADSCs) for treating various neurological disorders have been conducted and published (see Table 5). However, most of the studies have only demonstrated the safety hMSCs administration. Although the therapeutic effects have been seen in many clinical trials, the number of patients in each trial is usually not sufficient for analyzing the efficacy. Therefore, it is necessary to take hMSCs to the next step using more patients and proper controls to study their therapeutic effects and the mode of action in specific neurological disorders.

3.6. Dental Pulp Stem Cells (DPSCs) and Stem Cells from Human Exfoliated Deciduous Teeth (SHED)

DPSCs and SHED are derived from the dental pulp of adult permanent teeth and baby deciduous teeth, respectively. They are ectoderm-derived stem cells originating from neural crest cells and possess similar characteristics as MSCs, including the capacity of self-renew and multi-lineage differentiation and expression of MSC-related markers. However, it is still controversial to define DPSCs and SHED as MSCs, mainly due to their different potency of differentiating into specific lineages [148]. Because of their easy accessibility by routine dental procedures and their MSC-like properties, DPSCs and SHED have gained more attention in the last decade in the field of regenerative medicine, including treating neurological diseases. Moreover, they can maintain their stemness and multipotency for many years by cryopreservation, therefore, providing an opportunity for cell banking [149,150].
Remarkably, it has been reported that, compared to BM-MSCs or hADSCs, DPSCs have a higher growth rate, stronger neurogenesis, and better neuro-supportive and neuro-protective properties in neurological injuries and pathologies [148], indicating they may have better therapeutic effects for neurological disorders. DPSCs have been used to treat many neurological disorders in animal studies and have led to significant beneficial outcomes (see Table S5), mainly through the secretion of neurotrophic factors and anti-inflammatory functions [151]. It has been reported that the expression of neurotrophic factors in DPSCs is higher than that of hADSCs and BM-MSCs [152]. The conditioned medium from DPSCs can also improve several neuropathological conditions, including ALS [151,153]. On the other hand, the therapeutic effects of SHED for neurological disorders have also been studied in some animal studies, which have shown improved results (see Table S5). It is suggested that SHED are in a more immature state than DPSCs with higher expression of pluripotent markers and a higher proliferation rate, while DPSCs show higher expression of neuroectodermal markers [154,155]. Similarly, the conditioned medium of SHED has also been proved to be therapeutic for animal model of stroke, PD, and TBI etc [156,157,158].
Overall, DPSCs and SHED are also promising candidates for treating neurological disorders and may be beyond hADSCs and BM-MSCs. However, due to the limited volume of the pulp tissue, it usually takes months to obtain enough cells for therapy from the primary isolation, although they have a high proliferation rate [148]. Moreover, the heterogeneity of DPSCs and SHED may affect their therapeutic potentials [159,160]. Moreover, most of the available evidence of their therapeutic function for neurological disorders was acquired using nonhuman xenotransplants. It is still a long way to study their therapeutic and side effects in humans.

3.7. Muse Cells

Muse cells (multilineage-differentiating stress enduring cells) are non-cancerous pluripotent stem cells that were first discovered by Yasumasa et al. in 2010 [161]. Muse cells are sporadically present in the connective tissue of nearly all organs, such as bone marrow and umbilical cord, and can even be collected from commercial cell lines, including human fibroblasts and bone marrow MSCs [161,162,163]. Interestingly, it has been reported that Muse cells are the primary source of hiPSCs in human fibroblasts, but not the non-Muse cells [164,165]. Moreover, as Muse cells are present in cultured MSCs, it is thought that Muse cells are more therapeutic in clinical MSCs therapies regarding tissue regeneration [166]. The characteristics of Muse cells, including non-tumorigenicity, pluripotency, and easy collection, have made Muse cells a promising candidate for cell therapy.
Several animal studies have shown the safety and effectiveness of human Muse cells for treating neurological disorders, such as stroke, intracerebral hemorrhage, encephalopathy, ALS, and SCI (see Table S6). Moreover, Muse cells can engraft and integrate into the damaged regions and differentiate into neuronal cells, and finally, lead to functional and morphological recovery after intravenous administration [167,168,169,170]. However, Muse cells are still a novel type of stem cells that have not been well studied as other stem cells. With more related studies, we will obtain a better understanding of Muse cells and their potentials for treating neurological disorders.

3.8. Olfactory Ensheathing Cells (OECs)

OECs, also known as olfactory ensheathing glia, are terminally differentiated and self-renewable cells that are found in both the peripheral nervous system (PNS) and CNS, supporting the regeneration of the olfactory system throughout life. OECs can be isolated from the olfactory bulb (OB-OECs) through intracranial surgery or from olfactory mucosa (OM-OECs) through a simple, non-invasive nasal biopsy, which is preferred for autologous transplantation. Based on these properties, human OECs (hOECs) are considered a suitable candidate for CNS transplantation, particularly for SCI treatment. Numerous animal studies have shown that hOECs could promote the recovery from SCI through various mechanisms, including neuroprotection, promoting axonal growth/sprouting, improving angiogenesis, and restriction of glial scar [171,172]. Notably, OECs from different sites may act through different mechanisms. It has been reported that OM-OECs could regulate extracellular matrix and improve angiogenesis, while OB-OECs intend to improve axonal regeneration [172]. In addition to the animal studies, hOECs have also been used in clinical trials for SCI since the early 2000s (see Table 6). Although with a relatively small size of patients and variable outcomes in different patients, hOECs transplantation has led to different levels of functional recovery in most cases, indicating their therapeutic roles for SCI. Moreover, hOECs have also been used for treating ALS and cerebral palsy patients and have resulted in functional improvements, suggesting their therapeutic potentials are not only for SCI but also for other neurological disorders [173,174].
However, several bottlenecks are impeding the therapeutic effects of hOECs. hOECs isolated from the olfactory bulb or olfactory mucosa usually contain contamination cells such as fibroblasts that may affect their effects and need to be purified [175,176,177]. Moreover, the difficulty to quickly expand, poor survival after suspension injection, limited migration, and phagocytosis are also considered the major hampers of hOECs treatments [175]. Although various methods have been used to optimize OECs transplantation, it is still far from being a mature treatment for neurological disorders, including SCI [175].

3.9. Hematopoietic Stem Cells (HSCs)

HSCs are multipotent cells that generate all types of blood cells and can be found in peripheral blood, bone marrow (which provides an opportunity for autologous transplantation), and umbilical cord blood. Human HSCs (hHSCs) are one of the earliest cell types that have been used for clinical transplantation for the treatment of certain cancerous diseases, with acceptable safety [185]. Due to their ability to generate new blood and immune cells, hHSCs, especially autologous hHSCs, have also been used to treat autoimmune diseases of the nervous system, including MS, in the last two decades [186]. Currently, thousands of MS patients have been treated with autologous hHSCs, and many of them have shown significant therapeutic outcomes. However, in comparison to other stem cells, hHSCs only work as a supportive blood product following chemotherapy to speed hematopoietic recovery rather than a single treatment for these diseases. Actually, “autologous hematopoietic stem cell transplantation (AHSCT)” has become a clinical term that contains several procedures, including (1) mobilization—releasing hHSCs from the bone marrow into peripheral blood, (2) harvesting—collecting hHSCs from the blood of the patient, (3) conditioning regimen—administration of cytotoxic chemotherapy, and (4) infusion—returning hHSCs to the patient by infusion into the veins. Overall, with more clinical experience, AHSCT has become a promising supportive strategy for treating MS. The current status of using AHSCT for MS is well reviewed in a published paper [187].

4. Discussion

Cell therapy for neurological disorders has attracted more and more attention from researchers due to its unique and promising therapeutic potentials through the capabilities of cell replacement, neuroprotection, and promotion of intrinsic neuro-restoration. In this paper, we have introduced some promising cell types and their applications in treating neurological disorders both in animal studies and clinical trials. As we have summarized (see Table 7), each type of cell has its unique properties and may have different therapeutic functions in treating different diseases. However, it is worth mentioning that, although we have listed some common neurological disorders, the potential targets of the cells are not limited to what we have mentioned above. For example, MSCs have also been shown to be therapeutic for autism and meningitis, etc. [188,189]. An ideal cell type is not only about its therapeutic effects but also about its accessibility and cost and the time to obtain sufficient quantities. Therefore, it is hard to identify the best cell candidate for neurological disorders at the current stage without enough comparative data.
Aside from cell type, cell source, and the target disease, the outcome of cell therapy can also be highly influenced by other factors, including the route and cell quantity of cell administration. Intracerebral and intraspinal administration can bring more cells to the target position but may cause unexpected adverse effects due to the procedure, while intravenous or intranasal injections are safer but may lead to a loss of the cells. In addition, a successful cell therapy usually needs millions of cells for transplantation to humans; however, the cells are not “the more, the better” as more cells may result in negative outcomes, for example, reducing the safety of the cell therapy. Therefore, a proper delivery approach and a safe and therapeutic range of the quantity of the cells need to be found for specific diseases in future studies. Luckily, despite being at the pre-clinical stage of development, several methods have been proven to improve the therapeutic effects of the cells, such as genetic modification, pre-conditioning, and co-transplantation. With more studies conducted, it is possible to develop more effective cell therapies for patients with neurological disorders.
However, it is noteworthy that although animal studies usually show significant improvement after cell transplantation, the outcomes from clinical trials are normally not as suitable as from animal studies due to the differences between clinical diseases and animal models, such as the far greater heterogeneity between human patients than that of purpose-bred animals. Therefore, some success in animal studies may not be transferred to human studies, indicating the need to build up more clinically related animal models.

5. Conclusions

In this paper, we have introduced some promising cells that may be clinically therapeutic for the treatment of neurological disorders, based on their characteristics and the results from related clinical trials and recent animal studies. Overall, with more knowledge about these cells, it can be foreseen that cell therapy will have a crucial place in the future clinical management of neurological disorders, although there is still much work that remains to be done.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/biology10111142/s1, Table S1: Examples of hiPSC transplantation for neurological disorders in animal studies since 2016 (data from PubMed). Table S2: Examples of hNSPC transplantation for neurological disorders in animal studies since 2016 (data from PubMed). Table S3: Examples of human neuron, oligodendrocyte, and astrocyte transplantation for neurological disorders in animal studies since 2016 (data from PubMed). Table S4: Examples of hMSC transplantation for neurological disorders in animal studies since 2016 (data from PubMed). Table S5: Examples of DPSC and SHED transplantation for neurological disorders in animal studies since 2016 (data from PubMed). Table S6: Examples of human Muse cell transplantation for neurological disorders in animal studies since 2016 (data from PubMed).

Author Contributions

All authors contributed to the idea for this article. D.L. performed the literature search and original draft preparation. L.B. and X.-F.Z. critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Acknowledgments

We would like to thank the University of South Australia and the China Scholarship Council for their support throughout this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Muthuraman, M.; Koirala, N.; Ciolac, D.; Pintea, B.; Glaser, M.; Groppa, S.; Tamas, G.; Groppa, S. Deep brain stimulation and l-dopa therapy: Concepts of action and clinical applications in parkinson’s disease. Front. Neurol. 2018, 9, 711. [Google Scholar] [CrossRef] [PubMed]
  2. Matsunaga, S.; Kishi, T.; Iwata, N. Combination therapy with cholinesterase inhibitors and memantine for alzheimer’s disease: A systematic review and meta-analysis. Int. J. Neuropsychopharmacol. 2014, 18, P859–P860. [Google Scholar] [CrossRef]
  3. Besusso, D.; Schellino, R.; Boido, M.; Belloli, S.; Parolisi, R.; Conforti, P.; Faedo, A.; Cernigoj, M.; Campus, I.; Laporta, A.; et al. Stem cell-derived human striatal progenitors innervate striatal targets and alleviate sensorimotor deficit in a rat model of huntington disease. Stem Cell Rep. 2020, 14, 876–891. [Google Scholar] [CrossRef] [PubMed]
  4. Marques, C.R.; Marote, A.; Mendes-Pinheiro, B.; Teixeira, F.G.; Salgado, A.J. Cell secretome based approaches in parkinson’s disease regenerative medicine. Expert Opin. Biol. Ther. 2018, 18, 1235–1245. [Google Scholar] [CrossRef]
  5. Bellak, T.; Fekecs, Z.; Torok, D.; Tancos, Z.; Nemes, C.; Tezsla, Z.; Gal, L.; Polgari, S.; Kobolak, J.; Dinnyes, A.; et al. Grafted human induced pluripotent stem cells improve the outcome of spinal cord injury: Modulation of the lesion microenvironment. Sci. Rep. 2020, 10, 22414. [Google Scholar] [CrossRef]
  6. Mason, C.; Dunnill, P. Assessing the value of autologous and allogeneic cells for regenerative medicine. Regen. Med. 2009, 4, 835–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shroff, G. A review on stem cell therapy for multiple sclerosis: Special focus on human embryonic stem cells. Stem Cells Cloning 2018, 11, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. De Gioia, R.; Biella, F.; Citterio, G.; Rizzo, F.; Abati, E.; Nizzardo, M.; Bresolin, N.; Comi, G.P.; Corti, S. Neural stem cell transplantation for neurodegenerative diseases. Int. J. Mol. Sci. 2020, 21, 3103. [Google Scholar] [CrossRef] [PubMed]
  9. Zhou, T.; Benda, C.; Dunzinger, S.; Huang, Y.; Ho, J.C.; Yang, J.; Wang, Y.; Zhang, Y.; Zhuang, Q.; Li, Y.; et al. Generation of human induced pluripotent stem cells from urine samples. Nat. Protoc. 2012, 7, 2080–2089. [Google Scholar] [CrossRef] [PubMed]
  10. Capetian, P.; Azmitia, L.; Pauly, M.G.; Krajka, V.; Stengel, F.; Bernhardi, E.M.; Klett, M.; Meier, B.; Seibler, P.; Stanslowsky, N.; et al. Plasmid-based generation of induced neural stem cells from adult human fibroblasts. Front. Cell Neurosci. 2016, 10, 245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Augustyniak, J.; Zychowicz, M.; Podobinska, M.; Barta, T.; Buzanska, L. Reprogramming of somatic cells: Possible methods to derive safe, clinical-grade human induced pluripotent stem cells. Acta Neurobiol. Exp. 2014, 74, 373–382. [Google Scholar]
  12. Ebrahimi, A.; Keske, E.; Mehdipor, A.; Ebrahimi-Kalan, A.; Ghorbani, M. Somatic cell reprogramming as a tool for neurodegenerative diseases. Biomed. Pharmacother. 2019, 112, 108663. [Google Scholar] [CrossRef] [PubMed]
  13. Zhou, J.; Sun, J. A revolution in reprogramming: Small molecules. Curr. Mol. Med. 2019, 19, 77–90. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, W.Z.; Zhang, Y.; Wu, F.; Min, W.P.; Minev, B.; Zhang, M.; Luo, X.L.; Ramos, F.; Ichim, T.E.; Riordan, N.H.; et al. Safety evaluation of allogeneic umbilical cord blood mononuclear cell therapy for degenerative conditions. J. Transl. Med. 2010, 8, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lin, C.S.; Lin, G.; Lue, T.F. Allogeneic and xenogeneic transplantation of adipose-derived stem cells in immunocompetent recipients without immunosuppressants. Stem Cells Dev. 2012, 21, 2770–2778. [Google Scholar] [CrossRef] [Green Version]
  16. Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. McDonald, J.W.; Liu, X.Z.; Qu, Y.; Liu, S.; Mickey, S.K.; Turetsky, D.; Gottlieb, D.I.; Choi, D.W. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat. Med. 1999, 5, 1410–1412. [Google Scholar] [CrossRef]
  18. Srivastava, A.S.; Shenouda, S.; Mishra, R.; Carrier, E. Transplanted embryonic stem cells successfully survive, proliferate, and migrate to damaged regions of the mouse brain. Stem Cells 2006, 24, 1689–1694. [Google Scholar] [CrossRef]
  19. Liu, X.; Li, W.; Fu, X.; Xu, Y. The immunogenicity and immune tolerance of pluripotent stem cell derivatives. Front. Immunol. 2017, 8, 645. [Google Scholar] [CrossRef] [Green Version]
  20. Hentze, H.; Soong, P.L.; Wang, S.T.; Phillips, B.W.; Putti, T.C.; Dunn, N.R. Teratoma formation by human embryonic stem cells: Evaluation of essential parameters for future safety studies. Stem Cell Res. 2009, 2, 198–210. [Google Scholar] [CrossRef] [Green Version]
  21. Lee, A.S.; Tang, C.; Rao, M.S.; Weissman, I.L.; Wu, J.C. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat. Med. 2013, 19, 998–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Acquarone, M.; de Melo, T.M.; Meireles, F.; Brito-Moreira, J.; Oliveira, G.; Ferreira, S.T.; Castro, N.G.; Tovar-Moll, F.; Houzel, J.C.; Rehen, S.K. Mitomycin-treated undifferentiated embryonic stem cells as a safe and effective therapeutic strategy in a mouse model of parkinson’s disease. Front. Cell Neurosci. 2015, 9, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Matsuda, R.; Yoshikawa, M.; Kimura, H.; Ouji, Y.; Nakase, H.; Nishimura, F.; Nonaka, J.; Toriumi, H.; Yamada, S.; Nishiofuku, M.; et al. Cotransplantation of mouse embryonic stem cells and bone marrow stromal cells following spinal cord injury suppresses tumor development. Cell Transpl. 2009, 18, 39–54. [Google Scholar] [CrossRef] [PubMed]
  24. Erceg, S.; Ronaghi, M.; Oria, M.; Rosello, M.G.; Arago, M.A.; Lopez, M.G.; Radojevic, I.; Moreno-Manzano, V.; Rodriguez-Jimenez, F.J.; Bhattacharya, S.S.; et al. Transplanted oligodendrocytes and motoneuron progenitors generated from human embryonic stem cells promote locomotor recovery after spinal cord transection. Stem Cells 2010, 28, 1541–1549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Qiu, X.; Liu, Y.; Xiao, X.; He, J.; Zhang, H.; Li, Y. In vitro induction of human embryonic stem cells into the midbrain dopaminergic neurons and transplantation in cynomolgus monkey. Cell Reprogram. 2019, 21, 285–295. [Google Scholar] [CrossRef]
  26. de Castro, M.V.; da Silva, M.V.R.; Chiarotto, G.B.; Santana, M.H.A.; Luzo, A.C.M.; Kyrylenko, S.; de Oliveira, A.L.R. Spinal reflex recovery after dorsal rhizotomy and repair with platelet-rich plasma (prp) gel combined with bioengineered human embryonic stem cells (hescs). Stem Cells Int. 2020, 2020, 8834360. [Google Scholar] [CrossRef] [PubMed]
  27. Shroff, G. Evaluation of patients with multiple sclerosis using reverse nutech functional score and expanded disability status scale after human embryonic stem cell therapy. Clin. Transl. Med. 2016, 5, 43, Erratum in 2017, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  28. Shroff, G.; Thakur, D.; Dhingra, V.; Baroli, D.S.; Khatri, D.; Gautam, R.D. Expression of concern to: Role of physiotherapy in the mobilization of patients with spinal cord injury undergoing human embryonic stem cells transplantation. Clin. Transl. Med. 2017, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  29. Shroff, G. Single-photon emission tomography imaging in patients with lyme disease treated with human embryonic stem cells. Neuroradiol. J. 2018, 31, 157–167. [Google Scholar] [CrossRef]
  30. Shroff, G. Transplantation of human embryonic stem cells in patients with multiple sclerosis and lyme disease. Am. J. Case Rep. 2016, 17, 944–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Shroff, G.; Gupta, R. Human embryonic stem cells in the treatment of patients with spinal cord injury. Ann. Neurosci. 2015, 22, 208–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Shroff, G. Human embryonic stem cell therapy in chronic spinal cord injury: A retrospective study. Clin. Transl. Sci. 2016, 9, 168–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Shroff, G. Comparison of nutech functional score with european stroke scale for patients with cerebrovascular accident treated with human embryonic stem cells: Nfs for cva patients treated with hescs. J. Vasc. Interv. Neurol. 2017, 9, 35–43. [Google Scholar] [PubMed]
  34. Shroff, G.; Gupta, A.; Barthakur, J.K. Therapeutic potential of human embryonic stem cell transplantation in patients with cerebral palsy. J. Transl. Med. 2014, 12, 318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [Green Version]
  36. Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.H.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mrna. Cell Stem Cell 2010, 7, 618–630. [Google Scholar] [CrossRef] [Green Version]
  37. Okita, K.; Matsumura, Y.; Sato, Y.; Okada, A.; Morizane, A.; Okamoto, S.; Hong, H.; Nakagawa, M.; Tanabe, K.; Tezuka, K.; et al. A more efficient method to generate integration-free human ips cells. Nat. Methods 2011, 8, 409–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yu, J.; Chau, K.F.; Vodyanik, M.A.; Jiang, J.; Jiang, Y. Efficient feeder-free episomal reprogramming with small molecules. PLoS ONE 2011, 6, e17557. [Google Scholar] [CrossRef]
  39. Cherkashova, E.A.; Leonov, G.E.; Namestnikova, D.D.; Solov’eva, A.A.; Gubskii, I.L.; Bukharova, T.B.; Gubskii, L.V.; Goldstein, D.V.; Yarygin, K.N. Methods of generation of induced pluripotent stem cells and their application for the therapy of central nervous system diseases. Bull. Exp. Biol. Med. 2020, 168, 566–573. [Google Scholar] [CrossRef] [PubMed]
  40. Qin, J.; Gong, G.M.; Sun, S.L.; Qi, J.; Zhang, H.L.; Wang, Y.L.; Wang, N.; Wang, Q.M.; Ji, Y.; Gao, Y.; et al. Functional recovery after transplantation of induced pluripotent stem cells in a rat hemorrhagic stroke model. Neurosci. Lett. 2013, 554, 70–75. [Google Scholar] [CrossRef] [PubMed]
  41. Chang, D.J.; Lee, N.; Park, I.H.; Choi, C.; Jeon, I.; Kwon, J.; Oh, S.H.; Shin, D.A.; Do, J.T.; Lee, D.R.; et al. Therapeutic potential of human induced pluripotent stem cells in experimental stroke. Cell Transpl. 2013, 22, 1427–1440. [Google Scholar] [CrossRef]
  42. Palma-Tortosa, S.; Tornero, D.; Gronning Hansen, M.; Monni, E.; Hajy, M.; Kartsivadze, S.; Aktay, S.; Tsupykov, O.; Parmar, M.; Deisseroth, K.; et al. Activity in grafted human ips cell-derived cortical neurons integrated in stroke-injured rat brain regulates motor behavior. Proc. Natl. Acad. Sci. USA 2020, 117, 9094–9100. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, K.H.; Lin, K.C.; Wallace, C.G.; Li, Y.C.; Shao, P.L.; Chiang, J.Y.; Sung, P.H.; Yip, H.K. Human induced pluripotent stem cell-derived mesenchymal stem cell therapy effectively reduced brain infarct volume and preserved neurological function in rat after acute intracranial hemorrhage. Am. J. Transl. Res. 2019, 11, 6232–6248. [Google Scholar] [PubMed]
  44. Takahashi, J. Ips cell-based therapy for parkinson’s disease: A kyoto trial. Regen. Ther. 2020, 13, 18–22. [Google Scholar] [CrossRef] [PubMed]
  45. Tsuji, O.; Sugai, K.; Yamaguchi, R.; Tashiro, S.; Nagoshi, N.; Kohyama, J.; Iida, T.; Ohkubo, T.; Itakura, G.; Isoda, M.; et al. Concise review: Laying the groundwork for a first-in-human study of an induced pluripotent stem cell-based intervention for spinal cord injury. Stem Cells 2019, 37, 6–13. [Google Scholar] [CrossRef] [Green Version]
  46. Okubo, T.; Iwanami, A.; Kohyama, J.; Itakura, G.; Kawabata, S.; Nishiyama, Y.; Sugai, K.; Ozaki, M.; Iida, T.; Matsubayashi, K.; et al. Pretreatment with a gamma-secretase inhibitor prevents tumor-like overgrowth in human ipsc-derived transplants for spinal cord injury. Stem Cell Rep. 2016, 7, 649–663. [Google Scholar] [CrossRef] [Green Version]
  47. Ming, G.L.; Song, H. Adult neurogenesis in the mammalian central nervous system. Annu. Rev. Neurosci. 2005, 28, 223–250. [Google Scholar] [CrossRef]
  48. Kuhn, H.G.; Dickinson-Anson, H.; Gage, F.H. Neurogenesis in the dentate gyrus of the adult rat: Age-related decrease of neuronal progenitor proliferation. J. Neurosci. 1996, 16, 2027–2033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Alexanian, A.R. An efficient method for generation of neural-like cells from adult human bone marrow-derived mesenchymal stem cells. Regen. Med. 2010, 5, 891–900. [Google Scholar] [CrossRef]
  50. Kim, H.W.; Lee, H.S.; Kang, J.M.; Bae, S.H.; Kim, C.; Lee, S.H.; Schwarz, J.; Kim, G.J.; Kim, J.S.; Cha, D.H.; et al. Dual effects of human placenta-derived neural cells on neuroprotection and the inhibition of neuroinflammation in a rodent model of parkinson’s disease. Cell Transpl. 2018, 27, 814–830. [Google Scholar] [CrossRef] [Green Version]
  51. Zou, Y.; Zhao, Y.; Xiao, Z.; Chen, B.; Ma, D.; Shen, H.; Gu, R.; Dai, J. Comparison of regenerative effects of transplanting three-dimensional longitudinal scaffold loaded-human mesenchymal stem cells and human neural stem cells on spinal cord completely transected rats. ACS Biomater. Sci. Eng. 2020, 6, 1671–1680. [Google Scholar] [CrossRef]
  52. Lin, G.Q.; He, X.F.; Liang, F.Y.; Guo, Y.; Sunnassee, G.; Chen, J.; Cao, X.M.; Chen, Y.Y.; Pan, G.J.; Pei, Z.; et al. Transplanted human neural precursor cells integrate into the host neural circuit and ameliorate neurological deficits in a mouse model of traumatic brain injury. Neurosci. Lett. 2018, 674, 11–17. [Google Scholar] [CrossRef] [PubMed]
  53. Lu, M.H.; Ji, W.L.; Chen, H.; Sun, Y.Y.; Zhao, X.Y.; Wang, F.; Shi, Y.; Hu, Y.N.; Liu, B.X.; Wu, J.W.; et al. Intranasal transplantation of human neural stem cells ameliorates alzheimer’s disease-like pathology in a mouse model. Front. Aging Neurosci. 2021, 13, 10151. [Google Scholar] [CrossRef]
  54. Lee, I.S.; Jung, K.; Kim, I.S.; Lee, H.; Kim, M.; Yun, S.; Hwang, K.; Shin, J.E.; Park, K.I. Human neural stem cells alleviate alzheimer-like pathology in a mouse model. Mol. Neurodegener. 2015, 10, 38. [Google Scholar] [CrossRef] [Green Version]
  55. Liu, A.M.; Kang, S.; Yu, P.P.; Shi, L.L.; Zhou, L.B. Transplantation of human urine-derived neural progenitor cells after spinal cord injury in rats. Neurosci. Lett. 2020, 735, 135201. [Google Scholar] [CrossRef]
  56. Mendes-Pinheiro, B.; Teixeira, F.G.; Anjo, S.I.; Manadas, B.; Behie, L.A.; Salgado, A.J. Secretome of undifferentiated neural progenitor cells induces histological and motor improvements in a rat model of parkinson’s disease. Stem Cell Transl. Med. 2018, 7, 829–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kim, H.S.; Jeon, I.; Noh, J.E.; Lee, H.; Hong, K.S.; Lee, N.; Pei, Z.; Song, J. Intracerebral transplantation of bdnf-overexpressing human neural stem cells (hb1.F3.Bdnf) promotes migration, differentiation and functional recovery in a rodent model of huntington’s disease. Exp. Neurobiol. 2020, 29, 130–137. [Google Scholar] [CrossRef] [PubMed]
  58. Thomsen, G.M.; Avalos, P.; Ma, A.A.; Alkaslasi, M.; Cho, N.; Wyss, L.; Vit, J.P.; Godoy, M.; Suezaki, P.; Shelest, O.; et al. Transplantation of neural progenitor cells expressing glial cell line-derived neurotrophic factor into the motor cortex as a strategy to treat amyotrophic lateral sclerosis. Stem Cells 2018, 36, 1122–1131. [Google Scholar] [CrossRef] [Green Version]
  59. McGinley, L.M.; Kashlan, O.N.; Bruno, E.S.; Chen, K.S.; Hayes, J.M.; Kashlan, S.R.; Raykin, J.; Johe, K.; Murphy, G.G.; Feldman, E.L. Human neural stem cell transplantation improves cognition in a murine model of alzheimer’s disease. Sci. Rep. 2018, 8, 14776. [Google Scholar] [CrossRef]
  60. Okubo, T.; Nagoshi, N.; Kohyama, J.; Tsuji, O.; Shinozaki, M.; Shibata, S.; Kase, Y.; Matsumoto, M.; Nakamura, M.; Okano, H. Treatment with a gamma-secretase inhibitor promotes functional recovery in human ipsc- derived transplants for chronic spinal cord injury. Stem Cell Rep. 2018, 11, 1416–1432. [Google Scholar] [CrossRef] [Green Version]
  61. Ould-Brahim, F.; Sarma, S.N.; Syal, C.; Lu, K.J.; Seegobin, M.; Carter, A.; Jeffers, M.S.; Dor, C.; Stanford, W.L.; Corbett, D.; et al. Metformin preconditioning of human induced pluripotent stem cell-derived neural stem cells promotes their engraftment and improves post-stroke regeneration and recovery. Stem Cells Dev. 2018, 27, 1085–1096. [Google Scholar] [CrossRef]
  62. Kim, M.; Jung, K.; Ko, Y.; Kim, I.S.; Hwang, K.; Jang, J.H.; Shin, J.E.; Park, K.I. Tnf-alpha pretreatment improves the survival and function of transplanted human neural progenitor cells following hypoxic-ischemic brain injury. Cells 2020, 9, 1195. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, L.; Wang, F.; Chen, H.; Liu, D.; Qu, T.; Li, X.; Xu, D.; Liu, F.; Yin, Z.; Chen, Y. Co-transplantation of human umbilical cord mesenchymal stem cells and human neural stem cells improves the outcome in rats with spinal cord injury. Cell Transpl. 2019, 28, 893–906. [Google Scholar] [CrossRef] [Green Version]
  64. Zou, Y.L.; Ma, D.Z.; Shen, H.; Zhao, Y.N.; Xu, B.; Fan, Y.H.; Sun, Z.; Chen, B.; Xue, W.W.; Shi, Y.; et al. Aligned collagen scaffold combination with human spinal cord-derived neural stem cells to improve spinal cord injury repair. Biomater. Sci. 2020, 8, 5145–5156. [Google Scholar] [CrossRef]
  65. Abdolahi, S.; Aligholi, H.; Khodakaram-Tafti, A.; Ghadiri, M.K.; Stummer, W.; Gorji, A. Improvement of rat spinal cord injury following lentiviral vector-transduced neural stem/progenitor cells derived from human epileptic brain tissue transplantation with a self-assembling peptide scaffold. Mol. Neurobiol. 2021, 58, 2481–2493. [Google Scholar] [CrossRef]
  66. Kajikawa, K.; Imaizumi, K.; Shinozaki, M.; Shibata, S.; Shindo, T.; Kitagawa, T.; Shibata, R.; Kamata, Y.; Kojima, K.; Nagoshi, N.; et al. Cell therapy for spinal cord injury by using human ipsc-derived region-specific neural progenitor cells. Mol. Brain 2020, 13, 120. [Google Scholar] [CrossRef] [PubMed]
  67. Feldman, E.L.; Boulis, N.M.; Hur, J.; Johe, K.; Rutkove, S.B.; Federici, T.; Polak, M.; Bordeau, J.; Sakowski, S.A.; Glass, J.D. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: Phase 1 trial outcomes. Ann. Neurol. 2014, 75, 363–373. [Google Scholar] [CrossRef] [Green Version]
  68. Curtis, E.; Martin, J.R.; Gabel, B.; Sidhu, N.; Rzesiewicz, T.K.; Mandeville, R.; Van Gorp, S.; Leerink, M.; Tadokoro, T.; Marsala, S.; et al. A first-in-human, phase i study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell 2018, 22, 941–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Zhang, G.Z.; Li, Y.; Reuss, J.L.; Liu, N.; Wu, C.Y.; Li, J.P.; Xu, S.S.; Wang, F.; Hazel, T.G.; Cunningham, M.; et al. Stable intracerebral transplantation of neural stem cells for the treatment of paralysis due to ischemic stroke. Stem Cell Transl. Med. 2019, 8, 999–1007. [Google Scholar] [CrossRef] [Green Version]
  70. Chen, G.J.; Wang, Y.L.; Xu, Z.Y.; Fang, F.; Xu, R.M.; Wang, Y.; Hu, X.L.; Fan, L.X.; Liu, H.Q. Neural stem cell-like cells derived from autologous bone mesenchymal stem cells for the treatment of patients with cerebral palsy. J. Transl. Med. 2013, 11, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Harris, V.K.; Vyshkina, T.; Sadiq, S.A. Clinical safety of intrathecal administration of mesenchymal stromal cell-derived neural progenitors in multiple sclerosis. Cytotherapy 2016, 18, 1476–1482. [Google Scholar] [CrossRef]
  72. Wang, Z.G.; Luo, Y.; Chen, L.A.; Liang, W. Safety of neural stem cell transplantation in patients with severe traumatic brain injury. Exp. Ther. Med. 2017, 13, 3613–3618. [Google Scholar] [CrossRef] [Green Version]
  73. Harris, V.K.; Stark, J.; Vyshkina, T.; Blackshear, L.; Joo, G.; Stefanova, V.; Sara, G.; Sadiq, S.A. Phase i trial of intrathecal mesenchymal stem cell-derived neural progenitors in progressive multiple sclerosis. Ebiomedicine 2018, 29, 23–30. [Google Scholar] [CrossRef] [Green Version]
  74. Erharter, A.; Rizzi, S.; Mertens, J.; Edenhofer, F. Take the shortcut—direct conversion of somatic cells into induced neural stem cells and their biomedical applications. FEBS Lett. 2019, 593, 3353–3369. [Google Scholar] [CrossRef]
  75. Liu, D.H.; Manaph, N.P.A.; Al-Hawwas, M.; Zhou, X.F.; Liao, H. Small molecules for neural stem cell induction. Stem Cells Dev. 2018, 27, 297–312. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Z.; Zhao, W.; Liu, W.; Zhou, Y.; Jia, J.; Yang, L. Transplantation of placenta-derived mesenchymal stem cell-induced neural stem cells to treat spinal cord injury. Neural Regen. Res. 2014, 9, 2197–2204. [Google Scholar]
  77. Bago, J.R.; Alfonso-Pecchio, A.; Okolie, O.; Dumitru, R.; Rinkenbaugh, A.; Baldwin, A.S.; Miller, C.R.; Magness, S.T.; Hingtgen, S.D. Therapeutically engineered induced neural stem cells are tumour-homing and inhibit progression of glioblastoma. Nat. Commun. 2016, 7, 10593. [Google Scholar] [CrossRef] [Green Version]
  78. Yamashita, T.; Liu, W.; Matsumura, Y.; Miyagi, R.; Zhai, Y.; Kusaki, M.; Hishikawa, N.; Ohta, Y.; Kim, S.M.; Kwak, T.H.; et al. Novel therapeutic transplantation of induced neural stem cells for stroke. Cell Transpl. 2017, 26, 461–467. [Google Scholar] [CrossRef] [Green Version]
  79. Vonderwalde, I.; Azimi, A.; Rolvink, G.; Ahlfors, J.E.; Shoichet, M.S.; Morshead, C.M. Transplantation of directly reprogrammed human neural precursor cells following stroke promotes synaptogenesis and functional recovery. Transl. Stroke Res. 2020, 11, 93–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Mazzini, L.; Gelati, M.; Profico, D.C.; Soraru, G.; Ferrari, D.; Copetti, M.; Muzi, G.; Ricciolini, C.; Carletti, S.; Giorgi, C.; et al. Results from phase i clinical trial with intraspinal injection of neural stem cells in amyotrophic lateral sclerosis: A long-term outcome. Stem Cell Transl. Med. 2019, 8, 887–897. [Google Scholar] [CrossRef] [Green Version]
  81. Glass, J.D.; Hertzberg, V.S.; Boulis, N.M.; Riley, J.; Federici, T.; Polak, M.; Bordeau, J.; Fournier, C.; Johe, K.; Hazel, T.; et al. Transplantation of spinal cord-derived neural stem cells for als: Analysis of phase 1 and 2 trials. Neurology 2016, 87, 392–400. [Google Scholar] [CrossRef] [Green Version]
  82. Madrazo, I.; Kopyov, O.; Avila-Rodriguez, M.A.; Ostrosky, F.; Carrasco, H.; Kopyov, A.; Avendano-Estrada, A.; Jimenez, F.; Magallon, E.; Zamorano, C.; et al. Transplantation of human neural progenitor cells (npc) into putamina of parkinsonian patients: A case series study, safety and efficacy four years after surgery. Cell Transpl. 2019, 28, 269–285. [Google Scholar] [CrossRef]
  83. Levi, A.D.; Anderson, K.D.; Okonkwo, D.O.; Park, P.; Bryce, T.N.; Kurpad, S.N.; Aarabi, B.; Hsieh, J.; Gant, K. Clinical outcomes from a multi-center study of human neural stem cell transplantation in chronic cervical spinal cord injury. J. Neurotraum. 2019, 36, 891–902. [Google Scholar] [CrossRef] [PubMed]
  84. Levi, A.D.; Okonkwo, D.O.; Park, P.; Jenkins, A.L.; Kurpad, S.N.; Parr, A.M.; Ganju, A.; Aarabi, B.; Kim, D.; Casha, S.; et al. Emerging safety of intramedullary transplantation of human neural stem cells in chronic cervical and thoracic spinal cord injury. Neurosurgery 2018, 82, 562–575. [Google Scholar] [CrossRef]
  85. Shin, J.C.; Kim, K.N.; Yoo, J.; Kim, I.S.; Yun, S.; Lee, H.; Jung, K.; Hwang, K.; Kim, M.; Lee, I.S.; et al. Clinical trial of human fetal brain-derived neural stem/progenitor cell transplantation in patients with traumatic cervical spinal cord injury. Neural. Plast. 2015, 2015, 630932. [Google Scholar] [CrossRef] [Green Version]
  86. Abeysinghe, H.C.S.; Bokhari, L.; Quigley, A.; Choolani, M.; Chan, J.; Dusting, G.J.; Crook, J.M.; Kobayashi, N.R.; Roulston, C.L. Pre-differentiation of human neural stem cells into gabaergic neurons prior to transplant results in greater repopulation of the damaged brain and accelerates functional recovery after transient ischemic stroke. Stem Cell Res. Ther. 2015, 6, 186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Jonsson, M.E.; Ono, Y.; Bjorklund, A.; Thompson, L.H. Identification of transplantable dopamine neuron precursors at different stages of midbrain neurogenesis. Exp. Neurol. 2009, 219, 341–354. [Google Scholar] [CrossRef] [PubMed]
  88. Watmuff, B.; Pouton, C.W.; Haynes, J.M. In vitro maturation of dopaminergic neurons derived from mouse embryonic stem cells: Implications for transplantation. PLoS ONE 2012, 7, e31999. [Google Scholar] [CrossRef] [Green Version]
  89. Payne, S.L.; Anandakumaran, P.N.; Varga, B.V.; Morshead, C.M.; Nagy, A.; Shoichet, M.S. In vitro maturation of human ipsc-derived neuroepithelial cells influences transplant survival in the stroke-injured rat brain. Tissue Eng. Part A 2018, 24, 351–360. [Google Scholar] [CrossRef]
  90. Fortin, J.M.; Azari, H.; Zheng, T.; Darioosh, R.P.; Schmoll, M.E.; Vedam-Mai, V.; Deleyrolle, L.P.; Reynolds, B.A. Transplantation of defined populations of differentiated human neural stem cell progeny. Sci. Rep. 2016, 6, 23579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Qiu, L.; Liao, M.C.; Chen, A.K.; Wei, S.; Xie, S.; Reuveny, S.; Zhou, Z.D.; Hunziker, W.; Tan, E.K.; Oh, S.K.W.; et al. Immature midbrain dopaminergic neurons derived from floor-plate method improve cell transplantation therapy efficacy for parkinson’s disease. Stem Cells Transl. Med. 2017, 6, 1803–1814. [Google Scholar] [CrossRef] [PubMed]
  92. Lee, H.; Lee, H.Y.; Lee, B.E.; Gerovska, D.; Park, S.Y.; Zaehres, H.; Arauzo-Bravo, M.J.; Kim, J.I.; Ha, Y.; Scholer, H.R.; et al. Sequentially induced motor neurons from human fibroblasts facilitate locomotor recovery in a rodent spinal cord injury model. Elife 2020, 9, e52069. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, D.H.; Rychkov, G.; Al-Hawwas, M.; ul Manaph, N.P.A.; Zhou, F.N.; Bobrovskaya, L.; Liao, H.; Zhou, X.F. Conversion of human urine-derived cells into neuron-like cells by small molecules. Mol. Biol. Rep. 2020, 47, 2713–2722. [Google Scholar] [CrossRef]
  94. Salikhova, D.; Bukharova, T.; Cherkashova, E.; Namestnikova, D.; Leonov, G.; Nikitina, M.; Gubskiy, I.; Akopyan, G.; Elchaninov, A.; Midiber, K.; et al. Therapeutic effects of hipsc-derived glial and neuronal progenitor cells-conditioned medium in experimental ischemic stroke in rats. Int. J. Mol. Sci. 2021, 22, 4694. [Google Scholar] [CrossRef]
  95. Thiruvalluvan, A.; Czepiel, M.; Kap, Y.A.; Mantingh-Otter, I.; Vainchtein, I.; Kuipers, J.; Bijlard, M.; Baron, W.; Giepmans, B.; Bruck, W.; et al. Survival and functionality of human induced pluripotent stem cell-derived oligodendrocytes in a nonhuman primate model for multiple sclerosis. Stem Cells Transl. Med. 2016, 5, 1550–1561. [Google Scholar] [CrossRef] [Green Version]
  96. Fuhrmann, T.; Tam, R.Y.; Ballarin, B.; Coles, B.; Elliott Donaghue, I.; van der Kooy, D.; Nagy, A.; Tator, C.H.; Morshead, C.M.; Shoichet, M.S. Injectable hydrogel promotes early survival of induced pluripotent stem cell-derived oligodendrocytes and attenuates longterm teratoma formation in a spinal cord injury model. Biomaterials 2016, 83, 23–36. [Google Scholar] [CrossRef]
  97. Kawabata, S.; Takano, M.; Numasawa-Kuroiwa, Y.; Itakura, G.; Kobayashi, Y.; Nishiyama, Y.; Sugai, K.; Nishimura, S.; Iwai, H.; Isoda, M.; et al. Grafted human ips cell-derived oligodendrocyte precursor cells contribute to robust remyelination of demyelinated axons after spinal cord injury. Stem Cell Rep. 2016, 6, 1–8. [Google Scholar] [CrossRef] [Green Version]
  98. Keirstead, H.S.; Blakemore, W.F. Identification of post-mitotic oligodendrocytes incapable of remyelination within the demyelinated adult spinal cord. J. Neuropathol. Exp. Neurol. 1997, 56, 1191–1201. [Google Scholar] [CrossRef] [Green Version]
  99. Targett, M.P.; Sussman, J.; Scolding, N.; O’Leary, M.T.; Compston, D.A.; Blakemore, W.F. Failure to achieve remyelination of demyelinated rat axons following transplantation of glial cells obtained from the adult human brain. Neuropathol. Appl. Neurobiol. 1996, 22, 199–206. [Google Scholar] [CrossRef]
  100. Albert, K.; Niskanen, J.; Kalvala, S.; Lehtonen, S. Utilising induced pluripotent stem cells in neurodegenerative disease research: Focus on glia. Int. J. Mol. Sci. 2021, 22, 4334. [Google Scholar] [CrossRef] [PubMed]
  101. Ricci, G.; Volpi, L.; Pasquali, L.; Petrozzi, L.; Siciliano, G. Astrocyte-neuron interactions in neurological disorders. J. Biol. Phys. 2009, 35, 317–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Silver, J.; Miller, J.H. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 2004, 5, 146–156. [Google Scholar] [CrossRef] [PubMed]
  103. Li, K.; Javed, E.; Scura, D.; Hala, T.J.; Seetharam, S.; Falnikar, A.; Richard, J.P.; Chorath, A.; Maragakis, N.J.; Wright, M.C.; et al. Human ips cell-derived astrocyte transplants preserve respiratory function after spinal cord injury. Exp. Neurol. 2015, 271, 479–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Davies, S.J.; Shih, C.H.; Noble, M.; Mayer-Proschel, M.; Davies, J.E.; Proschel, C. Transplantation of specific human astrocytes promotes functional recovery after spinal cord injury. PLoS ONE 2011, 6, e17328. [Google Scholar] [CrossRef] [Green Version]
  105. Nakao, N.; Shintani-Mizushima, A.; Kakishita, K.; Itakura, T. The ability of grafted human sympathetic neurons to synthesize and store dopamine: A potential mechanism for the clinical effect of sympathetic neuron autografts in patients with parkinson’s disease. Exp. Neurol. 2004, 188, 65–73. [Google Scholar] [CrossRef]
  106. Freed, C.R.; Greene, P.E.; Breeze, R.E.; Tsai, W.Y.; DuMouchel, W.; Kao, R.; Dillon, S.; Winfield, H.; Culver, S.; Trojanowski, J.Q.; et al. Transplantation of embryonic dopamine neurons for severe parkinson’s disease. N. Engl. J. Med. 2001, 344, 710–719. [Google Scholar] [CrossRef]
  107. Kondziolka, D.; Wechsler, L.; Goldstein, S.; Meltzer, C.; Thulborn, K.R.; Gebel, J.; Jannetta, P.; DeCesare, S.; Elder, E.M.; McGrogan, M.; et al. Transplantation of cultured human neuronal cells for patients with stroke. Neurology 2000, 55, 565–569. [Google Scholar] [CrossRef]
  108. Torrente, Y.; Polli, E. Mesenchymal stem cell transplantation for neurodegenerative diseases. Cell Transpl. 2008, 17, 1103–1113. [Google Scholar] [CrossRef] [Green Version]
  109. Asgari Taei, A.; Dargahi, L.; Nasoohi, S.; Hassanzadeh, G.; Kadivar, M.; Farahmandfar, M. The conditioned medium of human embryonic stem cell-derived mesenchymal stem cells alleviates neurological deficits and improves synaptic recovery in experimental stroke. J. Cell Physiol. 2021, 236, 1967–1979. [Google Scholar] [CrossRef]
  110. Zhang, C.; Zhang, C.; Xu, Y.; Li, C.; Cao, Y.; Li, P. Exosomes derived from human placenta-derived mesenchymal stem cells improve neurologic function by promoting angiogenesis after spinal cord injury. Neurosci. Lett. 2020, 739, 135399. [Google Scholar] [CrossRef]
  111. Yang, C.H.; Wang, G.J.; Ma, F.F.; Yu, B.Q.; Chen, F.C.; Yang, J.; Feng, J.J.; Wang, Q. Repeated injections of human umbilical cord blood-derived mesenchymal stem cells significantly promotes functional recovery in rabbits with spinal cord injury of two noncontinuous segments. Stem Cell Res. Ther. 2018, 9, 136. [Google Scholar] [CrossRef] [Green Version]
  112. Zhang, L.; Wang, L.M.; Chen, W.W.; Ma, Z.; Han, X.; Liu, C.M.; Cheng, X.; Shi, W.; Guo, J.J.; Qin, J.B.; et al. Neural differentiation of human wharton’s jelly-derived mesenchymal stem cells improves the recovery of neurological function after transplantation in ischemic stroke rats. Neural Regen. Res. 2017, 12, 1103–1110. [Google Scholar] [CrossRef]
  113. Lo Furno, D.; Mannino, G.; Giuffrida, R. Functional role of mesenchymal stem cells in the treatment of chronic neurodegenerative diseases. J. Cell Physiol. 2018, 233, 3982–3999. [Google Scholar] [CrossRef]
  114. Ruzicka, J.; Kulijewicz-Nawrot, M.; Rodrigez-Arellano, J.J.; Jendelova, P.; Sykova, E. Mesenchymal stem cells preserve working memory in the 3xtg-ad mouse model of alzheimer’s disease. Int. J. Mol. Sci. 2016, 17, 152. [Google Scholar] [CrossRef]
  115. Zhou, H.L.; Zhang, H.R.; Yan, Z.J.; Xu, R.X. Transplantation of human amniotic mesenchymal stem cells promotes neurological recovery in an intracerebral hemorrhage rat model. Biochem. Bioph. Res. Commun. 2016, 475, 202–208. [Google Scholar] [CrossRef]
  116. Huang, P.; Freeman, W.D.; Edenfield, B.H.; Brott, T.G.; Meschia, J.F.; Zubair, A.C. Safety and efficacy of intraventricular delivery of bone marrow-derived mesenchymal stem cells in hemorrhagic stroke model. Sci. Rep. 2019, 9, 5674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Tsai, P.J.; Yeh, C.C.; Huang, W.J.; Min, M.Y.; Huang, T.H.; Ko, T.L.; Huang, P.Y.; Chen, T.H.; Hsu, S.P.C.; Soong, B.W.; et al. Xenografting of human umbilical mesenchymal stem cells from wharton’s jelly ameliorates mouse spinocerebellar ataxia type 1. Transl. Neurodegener. 2019, 8, 29. [Google Scholar] [CrossRef] [PubMed]
  118. Kim, S.; Chang, K.A.; Kim, J.; Park, H.G.; Ra, J.C.; Kim, H.S.; Suh, Y.H. The preventive and therapeutic effects of intravenous human adipose-derived stem cells in alzheimer’s disease mice. PLoS ONE 2012, 7, e45757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Li, J.F.; Zhang, D.J.; Geng, T.C.; Chen, L.; Huang, H.Y.; Yin, H.L.; Zhang, Y.Z.; Lou, J.Y.; Cao, B.Z.; Wang, Y.L. The potential of human umbilical cord-derived mesenchymal stem cells as a novel cellular therapy for multiple sclerosis. Cell Transpl. 2014, 23, S113–S122. [Google Scholar] [CrossRef]
  120. Mangin, G.; Cogo, A.; Moisan, A.; Bonnin, P.; Maier, B.; Kubis, N.; Consortium, R. Intravenous administration of human adipose derived-mesenchymal stem cells is not efficient in diabetic or hypertensive mice subjected to focal cerebral ischemia. Front. Neurosci. 2019, 13, 718. [Google Scholar] [CrossRef] [Green Version]
  121. Mohammadi, A.; Maleki-Jamshid, A.; Milan, P.B.; Ebrahimzadeh, K.; Faghihi, F.; Joghataei, M.T. Intrahippocampal transplantation of undifferentiated human chorionic-derived mesenchymal stem cells does not improve learning and memory in the rat model of sporadic alzheimer disease. Curr. Stem Cell Res. Ther. 2019, 14, 184–190. [Google Scholar] [CrossRef] [PubMed]
  122. Das, M.; Mayilsamy, K.; Tang, X.L.; Han, J.Y.; Foran, E.; Willing, A.E.; Mohapatra, S.S.; Mohapatra, S. Pioglitazone treatment prior to transplantation improves the efficacy of human mesenchymal stem cells after traumatic brain injury in rats. Sci. Rep. 2019, 9, 13646. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, S.J.; Zhang, Q.; Li, M.; Gao, P.; Huang, K.; Beejadhursing, R.; Jiang, W.; Lei, T.; Zhu, M.X.; Shu, K. Gdnf promotes survival and therapeutic efficacy of human adipose-derived mesenchymal stem cells in a mouse model of parkinson’s disease. Cell Transpl. 2020, 29, 963689720908512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Guo, M.G.; Wu, L.X.; Song, Z.Y.; Yang, B. Enhancement of neural stem cell proliferation in rats with spinal cord injury by a combination of repetitive transcranial magnetic stimulation (rtms) and human umbilical cord blood mesenchymal stem cells (hucb-mscs). Med. Sci. Monit. 2020, 26, e924445. [Google Scholar] [CrossRef]
  125. Deng, W.S.; Yang, K.; Liang, B.; Liu, Y.F.; Chen, X.Y.; Zhang, S. Collagen/heparin sulfate scaffold combined with mesenchymal stem cells treatment for canines with spinal cord injury: A pilot feasibility study. J Orthop. Surg. 2021, 29, 23094990211012293. [Google Scholar] [CrossRef]
  126. Oshita, J.; Okazaki, T.; Mitsuhara, T.; Imura, T.; Nakagawa, K.; Otsuka, T.; Kurose, T.; Tamura, T.; Abiko, M.; Takeda, M.; et al. Early transplantation of human cranial bone-derived mesenchymal stem cells enhances functional recovery in ischemic stroke model rats. Neurol. Med. Chir. 2020, 60, 83–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Yamaguchi, S.; Horie, N.; Satoh, K.; Ishikawa, T.; Mori, T.; Maeda, H.; Fukuda, Y.; Ishizaka, S.; Hiu, T.; Morofuji, Y.; et al. Age of donor of human mesenchymal stem cells affects structural and functional recovery after cell therapy following ischaemic stroke. J. Cerebr. Blood Flow Met. 2018, 38, 1199–1212. [Google Scholar] [CrossRef] [PubMed]
  128. Ko, P.W.; Park, S.; Kang, K.; Lim, Y.H.; Kim, S.R.; Suk, K.; Kim, K.S.; Lee, H.W. Human allogeneic bone marrow-derived mesenchymal stem cell therapy for cerebellar ataxia: A case report. Medicina 2021, 57, 334. [Google Scholar] [CrossRef]
  129. Siwek, T.; Jezierska-Wozniak, K.; Maksymowicz, S.; Barczewska, M.; Sowa, M.; Badowska, W.; Maksymowicz, W. Repeat administration of bone marrow-derived mesenchymal stem cells for treatment of amyotrophic lateral sclerosis. Med. Sci. Monit. 2020, 26, e927484. [Google Scholar] [CrossRef]
  130. Yang, Y.; Pang, M.; Du, C.; Liu, Z.Y.; Chen, Z.H.; Wang, N.X.; Zhang, L.M.; Chen, Y.Y.; Mo, J.; Dong, J.W.; et al. Repeated subarachnoid administrations of allogeneic human umbilical cord mesenchymal stem cells for spinal cord injury: A phase 1/2 pilot study. Cytotherapy 2021, 23, 57–64. [Google Scholar] [CrossRef]
  131. Deng, W.S.; Ma, K.; Liang, B.; Liu, X.Y.; Xu, H.Y.; Zhang, J.; Shi, H.Y.; Sun, H.T.; Chen, X.Y.; Zhang, S. Collagen scaffold combined with human umbilical cord-mesenchymal stem cells transplantation for acute complete spinal cord injury. Neural Regen. Res. 2020, 15, 1686–1700. [Google Scholar]
  132. Phedy, P.; Djaja, Y.P.; Gatam, L.; Kusnadi, Y.; Wirawan, R.P.; Tobing, I.M.S.; Subakir, N.; Mappulilu, A.; Prawira, M.A.; Yauwenas, R.; et al. Motoric recovery after transplantation of bone marrow derived mesenchymal stem cells in chronic spinal cord injury: A case report. Am. J. Case Rep. 2019, 20, 1299–1304. [Google Scholar] [CrossRef]
  133. Xiao, Z.F.; Tang, F.W.; Zhao, Y.N.; Han, G.; Yin, N.; Li, X.; Chen, B.; Han, S.F.; Jiang, X.F.; Yun, C.; et al. Significant improvement of acute complete spinal cord injury patients diagnosed by a combined criteria implanted with neuroregen scaffolds and mesenchymal stem cells. Cell Transpl. 2018, 27, 907–915. [Google Scholar] [CrossRef] [Green Version]
  134. Hur, J.W.; Cho, T.H.; Park, D.H.; Lee, J.B.; Park, J.Y.; Chung, Y.G. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: A human trial. J. Spinal Cord. Med. 2016, 39, 655–664. [Google Scholar] [CrossRef]
  135. Gu, J.W.; Huang, L.; Zhang, C.; Wang, Y.; Zhang, R.B.; Tu, Z.L.; Wang, H.D.; Zhou, X.H.; Xiao, Z.S.; Liu, Z.G.; et al. Therapeutic evidence of umbilical cord-derived mesenchymal stem cell transplantation for cerebral palsy: A randomized, controlled trial. Stem Cell Res. Ther. 2020, 11, 43. [Google Scholar] [CrossRef] [PubMed]
  136. Okur, S.C.; Erdogan, S.; Demir, C.S.; Gunel, G.; Karaoz, E. The effect of umbilical cord-derived mesenchymal stem cell transplantation in a patient with cerebral palsy: A case report. Int. J. Stem Cells 2018, 11, 141–147. [Google Scholar] [CrossRef] [Green Version]
  137. Zhang, C.; Huang, L.; Gu, J.; Zhou, X. Therapy for cerebral palsy by human umbilical cord blood mesenchymal stem cells transplantation combined with basic rehabilitation treatment: A case report. Glob. Pediatr. Health 2015, 2, 2333794X15574091. [Google Scholar] [CrossRef] [Green Version]
  138. Kabatas, S.; Civelek, E.; Sezen, G.B.; Kaplan, N.; Savrunlu, E.C.; Cetin, E.; Diren, F.; KaraOz, E. Functional recovery after wharton’s jelly-derived mesenchymal stem cell administration in a patient with traumatic brain injury: A pilot study. Turk. Neurosurg. 2020, 30, 914–922. [Google Scholar] [CrossRef] [PubMed]
  139. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Kim, A.S.; Johnson, J.N.; Bates, D.; Poggio, G.; Case, C.; McGrogan, M.; et al. Two-year safety and clinical outcomes in chronic ischemic stroke patients after implantation of modified bone marrow-derived mesenchymal stem cells (sb623): A phase 1/2a study. J. Neurosurg. 2019, 131, 1462–1472. [Google Scholar] [CrossRef] [PubMed]
  140. Tsang, K.S.; Ng, C.P.S.; Zhu, X.L.; Wong, G.K.C.; Lu, G.; Ahuja, A.T.; Wong, K.S.L.; Ng, H.K.; Poon, W.S. Phase i/ii randomized controlled trial of autologous bone marrow-derived mesenchymal stem cell therapy for chronic stroke. World J. Stem Cells 2017, 9, 133–143. [Google Scholar] [CrossRef] [PubMed]
  141. Honmou, O.; Houkin, K.; Matsunaga, T.; Niitsu, Y.; Ishiai, S.; Onodera, R.; Waxman, S.G.; Kocsis, J.D. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain 2011, 134, 1790–1807. [Google Scholar] [CrossRef] [Green Version]
  142. Kabatas, S.; Civelek, E.; Inci, C.; Yalcinkaya, E.Y.; Gunel, G.; Kir, G.; Albayrak, E.; Ozturk, E.; Adas, G.; Karaoz, E. Wharton’s jelly-derived mesenchymal stem cell transplantation in a patient with hypoxic-ischemic encephalopathy: A pilot study. Cell Transpl. 2018, 27, 1425–1433. [Google Scholar] [CrossRef]
  143. Xie, B.C.; Gu, P.; Wang, W.T.; Dong, C.; Zhang, L.N.; Zhang, J.; Liu, H.M.; Qiu, F.C.; Han, R.; Zhang, Z.Q.; et al. Therapeutic effects of human umbilical cord mesenchymal stem cells transplantation on hypoxic ischemic encephalopathy. Am. J. Transl. Res. 2016, 8, 3241–3250. [Google Scholar]
  144. Riordan, N.H.; Morales, I.; Fernandez, G.; Allen, N.; Fearnot, N.E.; Leckrone, M.E.; Markovich, D.J.; Mansfield, D.; Avila, D.; Patel, A.N.; et al. Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cells in the treatment of multiple sclerosis. J. Transl. Med. 2018, 16, 57. [Google Scholar] [CrossRef] [Green Version]
  145. Cohen, J.A.; Imrey, P.B.; Planchon, S.M.; Bermel, R.A.; Fisher, E.; Fox, R.J.; Bar-Or, A.; Sharp, S.L.; Skaramagas, T.T.; Jagodnik, P.; et al. Pilot trial of intravenous autologous culture-expanded mesenchymal stem cell transplantation in multiple sclerosis. Mult. Scler. J. 2018, 24, 501–511. [Google Scholar] [CrossRef] [Green Version]
  146. Milczarek, O.; Jarocha, D.; Starowicz-Filip, A.; Kwiatkowski, S.; Badyra, B.; Majka, M. Multiple autologous bone marrow-derived cd271(+) mesenchymal stem cell transplantation overcomes drug-resistant epilepsy in children. Stem Cell Transl. Med. 2018, 7, 20–33. [Google Scholar] [CrossRef] [Green Version]
  147. Venkataramana, N.K.; Kumar, S.K.V.; Balaraju, S.; Radhakrishnan, R.C.; Bansal, A.; Dixit, A.; Rao, D.K.; Das, M.; Jan, M.; Gupta, P.K.; et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in parkinson’s disease. Transl. Res. 2010, 155, 62–70. [Google Scholar] [CrossRef] [PubMed]
  148. Lan, X.; Sun, Z.; Chu, C.; Boltze, J.; Li, S. Dental pulp stem cells: An attractive alternative for cell therapy in ischemic stroke. Front. Neurol. 2019, 10, 824. [Google Scholar] [CrossRef]
  149. Gioventu, S.; Andriolo, G.; Bonino, F.; Frasca, S.; Lazzari, L.; Montelatici, E.; Santoro, F.; Rebulla, P. A novel method for banking dental pulp stem cells. Transfus. Apher. Sci. 2012, 47, 199–206. [Google Scholar] [CrossRef] [PubMed]
  150. Ma, L.; Makino, Y.; Yamaza, H.; Akiyama, K.; Hoshino, Y.; Song, G.T.; Kukita, T.; Nonaka, K.; Shi, S.T.; Yamaza, T. Cryopreserved dental pulp tissues of exfoliated deciduous teeth is a feasible stem cell resource for regenerative medicine. PLoS ONE 2012, 7, e51777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Ueda, T.; Inden, M.; Ito, T.; Kurita, H.; Hozumi, I. Characteristics and therapeutic potential of dental pulp stem cells on neurodegenerative diseases. Front. Neurosci. 2020, 14, 407. [Google Scholar] [CrossRef] [PubMed]
  152. Mead, B.; Logan, A.; Berry, M.; Leadbeater, W.; Scheven, B.A. Paracrine-mediated neuroprotection and neuritogenesis of axotomised retinal ganglion cells by human dental pulp stem cells: Comparison with human bone marrow and adipose-derived mesenchymal stem cells. PLoS ONE 2014, 9, e109305. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, J.; Zuzzio, K.; Walker, C.L. Systemic dental pulp stem cell secretome therapy in a mouse model of amyotrophic lateral sclerosis. Brain Sci. 2019, 9, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Miura, M.; Gronthos, S.; Zhao, M.; Lu, B.; Fisher, L.W.; Robey, P.G.; Shi, S. Shed: Stem cells from human exfoliated deciduous teeth. Proc. Natl. Acad. Sci. USA 2003, 100, 5807–5812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Govindasamy, V.; Abdullah, A.N.; Ronald, V.S.; Musa, S.; Ab Aziz, Z.A.; Zain, R.B.; Totey, S.; Bhonde, R.R.; Abu Kasim, N.H. Inherent differential propensity of dental pulp stem cells derived from human deciduous and permanent teeth. J. Endod. 2010, 36, 1504–1515. [Google Scholar] [CrossRef] [Green Version]
  156. Inoue, T.; Sugiyama, M.; Hattori, H.; Wakita, H.; Wakabayashi, T.; Ueda, M. Stem cells from human exfoliated deciduous tooth-derived conditioned medium enhance recovery of focal cerebral ischemia in rats. Tissue Eng. Part A 2013, 19, 24–29. [Google Scholar] [CrossRef] [Green Version]
  157. Chen, Y.R.; Lai, P.L.; Chien, Y.; Lee, P.H.; Lai, Y.H.; Ma, H.I.; Shiau, C.Y.; Wang, K.C. Improvement of impaired motor functions by human dental exfoliated deciduous teeth stem cell-derived factors in a rat model of parkinson’s disease. Int. J. Mol. Sci. 2020, 21, 3807. [Google Scholar] [CrossRef] [PubMed]
  158. Li, Y.; Yang, Y.Y.; Ren, J.L.; Xu, F.; Chen, F.M.; Li, A. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia m1/m2 polarization in rats. Stem Cell Res. Ther. 2017, 8, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Alraies, A.; Waddington, R.J.; Sloan, A.J.; Moseley, R. Evaluation of dental pulp stem cell heterogeneity and behaviour in 3d type i collagen gels. Biomed. Res. Int. 2020, 2020, 3034727. [Google Scholar] [CrossRef] [PubMed]
  160. Kunimatsu, R.; Nakajima, K.; Awada, T.; Tsuka, Y.; Abe, T.; Ando, K.; Hiraki, T.; Kimura, A.; Tanimoto, K. Comparative characterization of stem cells from human exfoliated deciduous teeth, dental pulp, and bone marrow-derived mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2018, 501, 193–198. [Google Scholar] [CrossRef]
  161. Kuroda, Y.; Kitada, M.; Wakao, S.; Nishikawa, K.; Tanimura, Y.; Makinoshima, H.; Goda, M.; Akashi, H.; Inutsuka, A.; Niwa, A.; et al. Unique multipotent cells in adult human mesenchymal cell populations. Proc. Natl. Acad. Sci. USA 2010, 107, 8639–8643. [Google Scholar] [CrossRef] [Green Version]
  162. Leng, Z.; Sun, D.; Huang, Z.; Tadmori, I.; Chiang, N.; Kethidi, N.; Sabra, A.; Kushida, Y.; Fu, Y.S.; Dezawa, M.; et al. Quantitative analysis of ssea3+ cells from human umbilical cord after magnetic sorting. Cell Transpl. 2019, 28, 907–923. [Google Scholar] [CrossRef] [PubMed]
  163. Kuroda, Y.; Wakao, S.; Kitada, M.; Murakami, T.; Nojima, M.; Dezawa, M. Isolation, culture and evaluation of multilineage-differentiating stress-enduring (muse) cells. Nat. Protoc. 2013, 8, 1391–1415. [Google Scholar] [CrossRef]
  164. Wakao, S.; Kitada, M.; Kuroda, Y.; Shigemoto, T.; Matsuse, D.; Akashi, H.; Tanimura, Y.; Tsuchiyama, K.; Kikuchi, T.; Goda, M.; et al. Multilineage-differentiating stress-enduring (muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc. Natl. Acad. Sci. USA 2011, 108, 9875–9880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Byrne, J.A.; Nguyen, H.N.; Reijo Pera, R.A. Enhanced generation of induced pluripotent stem cells from a subpopulation of human fibroblasts. PLoS ONE 2009, 4, e7118. [Google Scholar] [CrossRef]
  166. Dezawa, M. Muse cells provide the pluripotency of mesenchymal stem cells: Direct contribution of muse cells to tissue regeneration. Cell Transpl. 2016, 25, 849–861. [Google Scholar] [CrossRef] [Green Version]
  167. Suzuki, T.; Sato, Y.; Kushida, Y.; Tsuji, M.; Wakao, S.; Ueda, K.; Imai, K.; Iitani, Y.; Shimizu, S.; Hida, H.; et al. Intravenously delivered multilineage-differentiating stress enduring cells dampen excessive glutamate metabolism and microglial activation in experimental perinatal hypoxic ischemic encephalopathy. J. Cereb. Blood Flow Metab. 2020, 41, 1707–1720. [Google Scholar] [CrossRef]
  168. Ozuru, R.; Wakao, S.; Tsuji, T.; Ohara, N.; Matsuba, T.; Amuran, M.Y.; Isobe, J.; Iino, M.; Nishida, N.; Matsumoto, S.; et al. Rescue from stx2-producing e. Coli-associated encephalopathy by intravenous injection of muse cells in nod-scid mice. Mol. Ther. 2020, 28, 100–118. [Google Scholar] [CrossRef] [Green Version]
  169. Yamashita, T.; Kushida, Y.; Wakao, S.; Tadokoro, K.; Nomura, E.; Omote, Y.; Takemoto, M.; Hishikawa, N.; Ohta, Y.; Dezawa, M.; et al. Therapeutic benefit of muse cells in a mouse model of amyotrophic lateral sclerosis. Sci. Rep. 2020, 10, 17102. [Google Scholar] [CrossRef]
  170. Kajitani, T.; Endo, T.; Iwabuchi, N.; Inoue, T.; Takahashi, Y.; Abe, T.; Niizuma, K.; Tominaga, T. Association of intravenous administration of human muse cells with deficit amelioration in a rat model of spinal cord injury. J. Neurosurg. Spine 2021, 34, 648–655. [Google Scholar] [CrossRef]
  171. Watzlawick, R.; Rind, J.; Sena, E.S.; Brommer, B.; Zhang, T.; Kopp, M.A.; Dirnagl, U.; Macleod, M.R.; Howells, D.W.; Schwab, J.M. Olfactory ensheathing cell transplantation in experimental spinal cord injury: Effect size and reporting bias of 62 experimental treatments: A systematic review and meta-analysis. PLoS Biol. 2016, 14, e1002468. [Google Scholar] [CrossRef]
  172. Ursavas, S.; Darici, H.; Karaoz, E. Olfactory ensheathing cells: Unique glial cells promising for treatments of spinal cord injury. J. Neurosci. Res. 2021, 99, 1579–1597. [Google Scholar] [CrossRef] [PubMed]
  173. Chen, L.; Chen, D.; Xi, H.; Wang, Q.; Liu, Y.; Zhang, F.; Wang, H.; Ren, Y.; Xiao, J.; Wang, Y.; et al. Olfactory ensheathing cell neurorestorotherapy for amyotrophic lateral sclerosis patients: Benefits from multiple transplantations. Cell Transpl. 2012, 21 (Suppl. 1), S65–S77. [Google Scholar] [CrossRef] [Green Version]
  174. Chen, L.; Huang, H.; Xi, H.; Xie, Z.; Liu, R.; Jiang, Z.; Zhang, F.; Liu, Y.; Chen, D.; Wang, Q.; et al. Intracranial transplant of olfactory ensheathing cells in children and adolescents with cerebral palsy: A randomized controlled clinical trial. Cell Transpl. 2010, 19, 185–191. [Google Scholar] [CrossRef] [PubMed]
  175. Gilmour, A.D.; Reshamwala, R.; Wright, A.A.; Ekberg, J.A.K.; St John, J.A. Optimizing olfactory ensheathing cell transplantation for spinal cord injury repair. J. Neurotrauma 2020, 37, 817–829. [Google Scholar] [CrossRef]
  176. Tabakow, P.; Jarmundowicz, W.; Czapiga, B.; Fortuna, W.; Miedzybrodzki, R.; Czyz, M.; Huber, J.; Szarek, D.; Okurowski, S.; Szewczyk, P.; et al. Transplantation of autologous olfactory ensheathing cells in complete human spinal cord injury. Cell Transpl. 2013, 22, 1591–1612. [Google Scholar] [CrossRef] [PubMed]
  177. Chen, L.; Huang, H.; Xi, H.; Zhang, F.; Liu, Y.; Chen, D.; Xiao, J. A prospective randomized double-blind clinical trial using a combination of olfactory ensheathing cells and schwann cells for the treatment of chronic complete spinal cord injuries. Cell Transpl. 2014, 23 (Suppl. 1), S35–S44. [Google Scholar] [CrossRef] [Green Version]
  178. Tabakow, P.; Raisman, G.; Fortuna, W.; Czyz, M.; Huber, J.; Li, D.; Szewczyk, P.; Okurowski, S.; Miedzybrodzki, R.; Czapiga, B.; et al. Functional regeneration of supraspinal connections in a patient with transected spinal cord following transplantation of bulbar olfactory ensheathing cells with peripheral nerve bridging. Cell Transpl. 2014, 23, 1631–1655. [Google Scholar] [CrossRef] [Green Version]
  179. Rao, Y.J.; Zhu, W.X.; Du, Z.Q.; Jia, C.X.; Du, T.X.; Zhao, Q.A.; Cao, X.Y.; Wang, Y.J. Effectiveness of olfactory ensheathing cell transplantation for treatment of spinal cord injury. Genet. Mol. Res. 2014, 13, 4124–4129. [Google Scholar] [CrossRef] [PubMed]
  180. Rao, Y.; Zhu, W.; Guo, Y.; Jia, C.; Qi, R.; Qiao, R.; Cao, D.; Zhang, H.; Cui, Z.; Yang, L.; et al. Long-term outcome of olfactory ensheathing cell transplantation in six patients with chronic complete spinal cord injury. Cell Transpl. 2013, 22 (Suppl. 1), S21–S25. [Google Scholar] [CrossRef]
  181. Rao, Y.; Zhu, W.; Liu, H.; Jia, C.; Zhao, Q.; Wang, Y. Clinical application of olfactory ensheathing cells in the treatment of spinal cord injury. J. Int. Med. Res. 2013, 41, 473–481. [Google Scholar] [CrossRef]
  182. Wu, J.; Sun, T.; Ye, C.; Yao, J.; Zhu, B.; He, H. Clinical observation of fetal olfactory ensheathing glia transplantation (oegt) in patients with complete chronic spinal cord injury. Cell Transpl. 2012, 21 (Suppl. 1), S33–S37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Huang, H.; Xi, H.; Chen, L.; Zhang, F.; Liu, Y. Long-term outcome of olfactory ensheathing cell therapy for patients with complete chronic spinal cord injury. Cell Transpl. 2012, 21 (Suppl. 1), S23–S31. [Google Scholar] [CrossRef] [PubMed]
  184. Huang, H.; Chen, L.; Wang, H.; Xiu, B.; Li, B.; Wang, R.; Zhang, J.; Zhang, F.; Gu, Z.; Li, Y.; et al. Influence of patients’ age on functional recovery after transplantation of olfactory ensheathing cells into injured spinal cord injury. Chin. Med. J. 2003, 116, 1488–1491. [Google Scholar]
  185. Copelan, E.A. Medical progress: Hematopoietic stem-cell transplantation. N. Engl. J. Med. 2006, 354, 1813–1826. [Google Scholar] [CrossRef]
  186. Burman, J.; Tolf, A.; Hagglund, H.; Askmark, H. Autologous haematopoietic stem cell transplantation for neurological diseases. J. Neurol. Neurosurg. Psychiatry 2018, 89, 147–155. [Google Scholar] [CrossRef] [PubMed]
  187. Mariottini, A.; De Matteis, E.; Muraro, P.A. Haematopoietic stem cell transplantation for multiple sclerosis: Current status. BioDrugs 2020, 34, 307–325. [Google Scholar] [CrossRef]
  188. Price, J. Cell therapy approaches to autism: A review of clinical trial data. Mol. Autism 2020, 11, 37. [Google Scholar] [CrossRef]
  189. Ahn, S.Y.; Chang, Y.S.; Kim, Y.E.; Sung, S.I.; Sung, D.K.; Park, W.S. Mesenchymal stem cells transplantation attenuates brain injury and enhances bacterial clearance in escherichia coli meningitis in newborn rats. Pediatr. Res. 2018, 84, 778–785. [Google Scholar] [CrossRef] [Green Version]
Table 1. Differences between autologous transplantation and allogeneic transplantation.
Table 1. Differences between autologous transplantation and allogeneic transplantation.
Cell SourceAdditional Invasive
Procedures
ImmunogenicityEthical
Issues
Cell
Availability
Cell Type
Autologous transplantationPatients themselvesYesNo immune rejectionNoLimited by autologous cell cultureLimited, depending on the patient him/herself
Allogeneic transplantationOther donorsNoActivated immune response, immunosuppressants requiredYes (when it involves the use of human embryos)Cryopreserved stocks, suitable for big amount cell preservationVarious, depending on the donors (e.g., ESCs and FSCs)
Table 2. Examples of hESC transplantation for neurological disorders in clinical trials (data from PubMed).
Table 2. Examples of hESC transplantation for neurological disorders in clinical trials (data from PubMed).
Disease Route of
Administration
Cell SourceCell AmountNumber of
Patients
Longest Follow-Up Time (after 1st Transplantation)Outcome/ConclusionRef.
Lyme diseaseIntramuscular, intravenous, and other supplemental routes Human embryo N/A598 weeks43 patients showed significant improvement, 12 patients showed moderate improvement, 4 patients exhibited mild improvement in their brain perfusion; no deterioration was found[29]
Lyme disease and multiple sclerosisIntramuscular, intravenous, and other supplemental routesHuman embryoN/A2N/APatients showed remarkable neurological functional and histological improvement; no adverse events were reported[30]
Spinal cord injuryIntramuscular, intravenous, and other supplemental routesHuman embryohundreds of millions of cells in total55 yearsAll patients showed neurological functional improvement, 3/5 showed improved American Spinal Injury Association score (ASIA); no adverse events were reported[31]
Spinal cord injuryIntramuscular, intravenous, and other supplemental routes Human embryohundreds of millions of cells in total226 N/A70% of patients improved by at least one ASIA grade after 3 phases of treatment; no adverse events were reported[32]
StrokeIntramuscular, intravenous, and other supplemental routesHuman embryohundreds of millions of cells in total24N/AA large population of patients saw significant improvement regarding Nutech Functional Score and European Stroke Scale; no adverse events were reported[33]
MultiplesclerosisIntramuscular, intravenous, and other supplemental routesHuman embryohundreds of millions of cells in total24Around 1 yearPatients showed an improvement in parameters associated with MS when evaluated with reverse nutech functional score but not with the expanded disability status scale; no adverse events were reported[27]
Cerebral palsyIntramuscular, intravenous, and other supplemental routes Human embryohundreds of millions of cells in total91N/AMost patients showed significant improvement in Gross Motor Function Classification Scores Expanded and Revised (GMFCS-E & R)[34]
Table 3. Examples of hNSPC transplantation for neurological disorders in clinical trials (data from PubMed).
Table 3. Examples of hNSPC transplantation for neurological disorders in clinical trials (data from PubMed).
Disease (Model)Route of AdministrationCell SourceCell AmountNumber of PatientsLongest Follow-Up Time (after 1st Transplantation)Outcome/ConclusionRef.
Ischemic strokeIntracerebralHuman fetal spinal cord, cell line: NSI-5661.2 × 107, 2.4 × 107, or 7.2 × 107924 monthsAll patients showed significant behavioral and histological improvements[69]
ALSIntraspinalHuman fetal brain2.25–4.6 × 1061851 monthsNo serious adverse effects. Some patients showed temporary subjective clinical improvement[80]
ALSIntraspinalHuman spinal cord2 to 16 million159 monthsIntraspinal transplantation of human spinal cord-derived neural stem cells can be safely accomplished at high doses[81]
ALSIntraspinalHuman fetal spinal cord, cell line: NSI-566RSC1.5 million1530 monthsThis NSPCs line can be safely transplanted into both lumbar and/or cervical human spinal cord segments[67]
moderate PDIntracerebralHuman fetal brain2 million74 yearsNo adverse effects; enhanced midbrain dopaminergic activity; minor neuropsychological changes; 6/7 showed improved motor function; 5/7 showed better response to medication [82]
Chronic cervical SCIIntraspinalCells were prepared and released by StemCells Inc.15 to 40 million16 1 yearCell transplantation was safe, feasible, and well tolerated.Trends toward improvement in motor function and spasticity were seen[83]
completethoracic SCIIntraspinalHuman fetal spinal cord, cell line: NSI-566N/A4 27 monthsNo serious adverse events; 3/4 showed early signs of potential efficacy[68]
Chronic cervical and thoracic SCIIntraspinalHuman fetal brain, cell line: HuCNS-SC20 to 40 million291 yearCell transplantation was safe and feasible using a manual injection technique [84]
Traumatic cervical SCIIntraspinalHuman fetal telencephalon1 × 108 341 yearNo serious adverse effects, 5/19 of treated patients showed functional recovery, 1/15 untreated patients showed functional recovery[85]
MSIntrathecalAutologous MSCs3 × 10720Around 1 yearNo serious adverse effects, improved median Expanded Disability Status Scale (EDSS), 70% and 50% of the subjects demonstrated improved muscle strength and bladder function, respectively[73]
MSIntrathecalAutologous bone marrow MSCs0.08–17.6 million68.9 yearsNo serious adverse events; 4/6 showed a measurable clinical improvement[71]
non-acute severe TBIIntravenous or intrathecalAutologous MSCs20 to 40 million106 monthsNo serious adverse events, 7/10 patients presented different degrees of improvement in neurological function[72]
Cerebral palsysubarachnoid cavityAutologous bone marrow MSCs1–2 × 107 606 monthsNo serious adverse events. Treated group showed significant motor function recovery but no significant increases in the language quotients[70]
Table 4. Examples of neuron transplantation for neurological disorders in clinical trials (data from PubMed).
Table 4. Examples of neuron transplantation for neurological disorders in clinical trials (data from PubMed).
Cell TypeDisease (Model)Route of AdministrationCell SourceCell AmountNumber of PatientsLongest Follow-Up Time (after 1st Transplantation)Outcome/ConclusionRef.
sympathetic neuronsPDIntracerebralAutologous sympathetic neuronsN/A436 months,Clinical evaluations showed that an increase in the duration of levodopa-induced ‘‘on’’ phase, and the percent time spent in ‘‘off’’ phase exhibited a 30–40% reduction as compared to the pre-grafting values[105]
Dopamine neuron-contained tissuePDIntracerebralHuman embryonic mesencephalic tissueN/A401 yearhuman embryonic dopamine neuron transplants survive in patients with severe Parkinson’s disease and result in some clinical benefit in younger but not in older patients[106]
neuronal cellsStrokeIntracerebralHuman teratocarcinomaN/A1218 monthsNo adverse cell-related serologic or imaging-defined effects. The total European Stroke Scale score improved in six patients (3 to 10 points), with a mean improvement of 2.9 points in all patients[107]
Table 5. Examples of hMSCs transplantation for neurological disorders in clinical trials (data from PubMed).
Table 5. Examples of hMSCs transplantation for neurological disorders in clinical trials (data from PubMed).
Disease (Model)Route of AdministrationCell SourceCell AmountNumber of PatientsLongest Follow-Up Time (after 1st Transplantation)Outcome/ConclusionRef.
Cerebellar ataxiaIntrathecalBone marrow, (cell line: CS20BR08)2 × 106/kg 110 monthsNo adverse events reported. Improved K-SARA (Korean version of the Scale for the Assessment and Rating of Ataxia) scores[128]
ALSIntrathecalAutologous bone marrow30 × 106814 monthsNo change in progression rate in patients with an inherently slow course, but some decreased progression rate in patients with an inherently rapid course[129]
SCIIntrathecalUmbilical cord4 × 106/kg14312 monthsNo serious adverse events reported. Significant improvements in neurological dysfunction and recovery of quality of life[130]
Acute complete SCIIntraspinalUmbilical cord40 million4012 monthsPromoted recovery of neurological function[131]
Chronic SCI Intradural and intravenousAutologous bone marrow6.6–7.6 × 10715 yearsNo complication or serious adverse effects, improved motoric function[132]
Acute complete SCIIntraspinalUmbilical cord4 × 10721 yearNo obvious adverse symptoms reported, the supraspinal control of movements below the injury was regained by functional NeuroRegen scaffolds implantation with hMSCs[133]
SCIIntrathecalAutologous adipose9 × 107148 monthsNo serious adverse events. Several patients showed mild improvements in neurological function[134]
Cerebral palsyIntravenousUmbilical cord4.5–5.5 × 1073913 monthshMSCs transplantation was safe and effective at improving the gross motor and comprehensive function of children with cerebral palsy when combined with rehabilitation[135]
Cerebral palsyIntrathecal and intravenousUmbilical cord8 × 106/kg118 monthsNo serious adverse effects reported. hMSCs transplantation improved functional recovery, combined with rehabilitation[136]
Cerebral palsyIntravenousUmbilical cord80 × 10715 yearshMSCs transplantation with basic rehabilitation improved the motor and comprehensive function. No serious adverse events [137]
TBIIntrathecal, intramuscular, and intravenousWharton’s jelly18 × l06 /kg112 monthsNo important negative effects were reported. The patients’ speech, cognitive, memory, and fine motor skills were improved[138]
Chronic ischemic strokeIntracerebralBone marrow, cell line: SB6232.5–10 × 1061824 monthsAll experienced at least 1 treatment-emergent adverse event. 7 experienced 9 serious adverse events, which resolved without sequelae. Improved clinical outcomes[139]
Chronic strokeIntravenousAutologous bone marrow5.3 × 105–2.9 × 106/kg960 weeksNo adverse event reported. Improved neurological functions and clinical outcomes[140]
StrokeIntravenousAutologous bone marrow0.6 to1.6 × 1081212 monthsNo significant adverse effects were found. Improved neurological function[141]
HIEIntrathecal, intramuscular, and intravenousWharton’s jelly12 × 106/kg112 monthsImproved neurological recovery[142]
HIEIntravenousUmbilical cord1 × 10822180 daysNo significant adverse effects were found. Markedly improved recovery of neurological function, cognition ability, emotional reaction, and extrapyramidal function[143]
MSIntravenousUmbilical cord14 × 107201 yearNo serious adverse events reported. Improved functional recovery[144]
MSIntravenousAutologous bone marrow1–2 × 106/kg246 monthsNo serious adverse effects reported. No substantial evidence of inhibition of disease activity, tissue repair, or recovery of function[145]
MSIntravenousUmbilical cord12 × 106/kg2312 monthsNo significant adverse effects were found. Improved neurological function[119]
Drug-resistant epilepsyIntrathecalAutologous bone marrow7.4–16 × 10742 yearsCD271+ hMSCs, combined with autologous bone marrow nucleated cells transplantation, showed no serious adverse events but considerable neurological and cognitive improvement[146]
PDIntracerebralAutologous bone marrowN/A736 monthsNo significant adverse effects were found. Several patients showed improved neurological function[147]
Table 6. Examples of OECs transplantation for neurological disorders in clinical trials (data from PubMed).
Table 6. Examples of OECs transplantation for neurological disorders in clinical trials (data from PubMed).
DiseaseRoute of AdministrationCell SourceCell AmountNumber of PatientsLongest Follow-Up Time (after 1st Transplantation)Outcome/Conclusion (Targeting Behavioral and Histological Change)Ref.
SCIIntraspinalAutologous olfactory bulb5 × 105119 monthsImproved neurological and histopathological recovery, no adverse effects were seen[178]
SCIIntraspinalHuman fetal olfactory bulbs1 × 10676 monthsNo serious adverse effects were seen. All treated patients showed functional improvement, 4/5 showed improvement in electrophysiological tests[177]
SCIIntraspinalHuman fetal olfactory bulbs2–5 × 106 158 weeksNo serious adverse effects were seen. 12/15 showed obvious spinal function improvement, and 3/15 had slight improvement[179]
SCIIntraspinalAutologous nasal mucosa1.8–21.2 × 10661 yearno adverse findings related to olfactory mucosa biopsy or transplantation. All treated patients showed improved functional recovery, 2/3 of treated patients showed improved American Spinal Injury Association class[176]
SCIIntraspinalHuman fetal olfactory bulbs2 × 106624 monthsNo clinical complications were observed. All patients showed improved neurofunctional recovery[180]
SCIIntraspinalAutologous olfactory mucosaNot mentioned824 monthsNo clinical complications were observed. All patients showed improved neurofunctional recovery, 3/8 showed returned substantial sensation and motor activity in various muscles, 2/8 showed restored bladder function[181]
SCIIntraspinalHuman fetal olfactory bulbs5 × 105111.5 yearsAll patients had no complications or deterioration of neurological conditions. Sensation and spasticity improved moderately. Locomotion recovery was minimal[182]
SCIIntraspinalolfactory bulbs5 × 1051085.3 yearsNo serious adverse effects were seen. Improve neurological functions. Sufficient rehabilitation most likely played a critical role[183]
SCIIntraspinalolfactory bulbs5 × 10517112 weeksOECs transplantation can improve the neurological function of spinal cord of SCI patients regardless of their ages[184]
ALSIntracranial and/or intraspinalHuman fetal olfactory bulbs1–2 × 106/treatment, 1–5 treatments507N/Amultiple doses of cellular therapy serve a positive role in the treatment of ALS[173]
Cerebral palsyIntracranialHuman fetal olfactory bulbs2 × 10614 6 monthsOECs transplantation is effective for functional improvement in children and adolescents with cerebral palsy, yet without obvious side effects[174]
Table 7. Characteristics of cell candidates for neurological disorders through direct transplantation.
Table 7. Characteristics of cell candidates for neurological disorders through direct transplantation.
Cell TypeStemnessAdvantageDisadvantageExamples of Targeted Neurological Disorders in Animal Studies Examples of Targeted Neurological Disorders in Clinical Trials
hESCsPluripotent Unlimited proliferationEthical issues; risk of immune rejection, risk of tumor formationSCILyme disease, MS, SCI, stroke, cerebral palsy
hiPSCsPluripotentNo ethical issues; applicable for autologous transplantation; high accessibilityRisk of tumor formation, unpredictable mutagenesisSCI, strokeN/A
hNSPCsMultipotent Neural lineage differentiation; low risk of tumor formation; multiple sourcesComparatively low proliferationSCI, HD, stroke, PD, AD, ataxia, TBI, ALSStroke, ALS, PD, SCI, MS, TBI, cerebral palsy
NeuronsTerminal cellsNo risk of tumor formation; no unexpected differentiation Poor survival after transplantationSCI, ALS, PD, ADPD, stroke
OligodendrocytesTerminal cellsNo risk of tumor formation; no unexpected differentiationPoor survival after transplantationMS, SCIN/A
AstrocytesTerminal cellsNo risk of tumor formation; no unexpected differentiationEffects highly depend on the subtype of the cells; not much studiedSCIN/A
hMSCsMultipotentApplicable for autologous transplantation; high accessibility; low risk of tumor formationLimited neural differentiation; effects may be not as suitable as hNSPCsSCI, PD, stroke, TBI, ALS, ataxia, MS, AD, epilepsySCI, PD, stroke, TBI, ALS, ataxia, MS, epilepsy, cerebral palsy, HIE
DPSCsMultipotentApplicable for autologous transplantation; high accessibility; low risk of tumor formationLimited neural differentiation; high heterogeneity; low number of cells from pulp tissueSCI, HD, ataxia, stroke, PDN/A
SHEDMultipotentHigh accessibility; low risk of tumor formationLimited neural differentiation; high heterogeneity; low number of cells from pulp tissueSCI, strokeN/A
Muse cellsPluripotentApplicable for autologous transplantation; high accessibility; non-tumorigenicityNot much studiedSCI, stroke, HIE, ALSN/A
hOECsTerminal cellsApplicable for autologous transplantation; high accessibility; non-tumorigenicity; no unexpected differentiationHard to purify; poor survival after transplantation; limited migration and phagocytosis; SCISCI, ALS, cerebral palsy
hHSCsMultipotentApplicable for autologous transplantation; high accessibilitySome risk of serious adverse effectsN/AMS
Abbreviation: SCI = spinal cord injury; MS = multiple sclerosis; HD = Huntington’s disease; TBI = traumatic brain injury; PD = Parkinson’s disease; AD = Alzheimer’s disease; ALS = amyotrophic lateral sclerosis; HIE = hypoxic-ischemic encephalopathy; N/A = not applicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, D.; Bobrovskaya, L.; Zhou, X.-F. Cell Therapy for Neurological Disorders: The Perspective of Promising Cells. Biology 2021, 10, 1142. https://doi.org/10.3390/biology10111142

AMA Style

Liu D, Bobrovskaya L, Zhou X-F. Cell Therapy for Neurological Disorders: The Perspective of Promising Cells. Biology. 2021; 10(11):1142. https://doi.org/10.3390/biology10111142

Chicago/Turabian Style

Liu, Donghui, Larisa Bobrovskaya, and Xin-Fu Zhou. 2021. "Cell Therapy for Neurological Disorders: The Perspective of Promising Cells" Biology 10, no. 11: 1142. https://doi.org/10.3390/biology10111142

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop