IMR Press / JIN / Volume 21 / Issue 2 / DOI: 10.31083/j.jin2102044
Open Access Review
Mesenchymal stromal cells: cell-based therapies for traumatic central nervous system injuries
Show Less
1 Department of Pediatrics, The University of Tokyo Hospital, 113-8655 Hongo, Bunkyo-Ku, Tokyo, Japan
2 Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo,108-8639 Shirokanedai, Minato-Ku, Tokyo, Japan
*Correspondence: takeo-m@ims.u-tokyo.ac.jp (Takeo Mukai)
Academic Editor: Masato Nakafuku
J. Integr. Neurosci. 2022, 21(2), 44; https://doi.org/10.31083/j.jin2102044
Submitted: 31 July 2021 | Revised: 11 August 2021 | Accepted: 24 August 2021 | Published: 18 March 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Traumatic central nervous system (CNS) injury often causes irreversible impairment, and new alternative therapies for the treatment of CNS injury and sequelae are expected to be developed. Recently, mesenchymal stromal cells (MSCs) have started being used as cell therapy for neurological disorders such as traumatic CNS injury based on their immunomodulatory, neuroprotective, and neurorestorative abilities. Based on the premise of basic research, numerous clinical trials using MSCs for the treatment of traumatic CNS injury have been performed, and the feasibility and efficacy of this therapy have been reported. In this review we aimed to shed light on the characteristics of MSCs and to discuss the basic and clinical research and recent progress in clinical studies using MSCs to treat various traumatic neurological injuries.

Keywords
Mesenchymal stromal cell
Traumatic central nervous system injury
Neurological disorders
Umbilical cord
Neurotrophic factors
1. Introduction

Traumatic central nervous system (CNS) injury often causes irreversible disorders. As for the mechanisms of the traumatic CNS injury, the initial injury is reported to cause necrosis and apoptosis of neural cells, followed by a secondary degeneration resulted from the apoptosis of undamaged neurons [1]. Basically there are two specific categories by which traumatic CNS injury occurs: traumatic brain injury (TBI) and spinal cord injury (SCI).

TBI and SCI are both caused by physical insults to the brain and spinal cord suddenly inflicted in situations such as traffic accidents, falls, and sporting activity. TBI is a primary cause of unexpected death and may induce serious sequalae [2]. On the other hand, SCI is also a severe traumatic insult of the CNS. Although there is a wide range in the occurrence rate, the total worldwide incidence of SCI has been reported to be 3.6–195.4 per million people [3]. These injuries trigger a neuroinflammatory reaction and disrupt neuroimmune communication, leading to serious deficits in sensorimotor functions; this may either lead to unexpected death or induce serious disabilities, motor and cognitive dysfunction [2]. TBI is also suspected to be a potential risk factor of neurodegenerative diseases, such as Alzheimer’s disease [4], amyotrophic lateral sclerosis [5], and Parkinson’s disease [6].

As for the treatment of traumatic CNS injuries, in addition to rehabilitative therapy, decompressive craniotomy, hyperosmolar treatment, and hypothermia therapy are performed to decrease intracranial pressure. However, these therapies are not perfect treatments in some cases. Moreover, most pharmacological trials that have been conducted and included glutamate antagonists, corticosteroids, free-radical scavengers, progesterone, have failed to demonstrate a significant clinical efficacy [7, 8]. Therefore, new therapeutic alternatives for the treatment of CNS injury and sequelae are expected to be developed and clinical trials of these new therapeutics are urgent issues.

Recently, mesenchymal stromal cells (MSCs), also called mesenchymal stem cells, are used to treat various diseases, and researchers have been focusing on their immunomodulatory and neurotrophic abilities. MSCs have been reported to be effective in neurological disorder models such as traumatic CNS models; additionally, clinical trials using MSCs for the treatment of traumatic CNS injuries have already been demonstrated, which are summarized in the present review.

In this paper, we characterized MSCs, especially umbilical cord (UC)-derived MSCs, presented methods of isolation and cryopreservation, and discussed their efficacy and mechanisms of action in treating traumatic CNS injuries, as well as their application in clinical trials.

2. Methods to harvest and cryopreserve UC-MSCs

Following many experiments, MSCs have been reported to be able to be harvested from various tissues, including bone marrow (BM) [9], UC blood (UCB) [10], adipose tissue (AD) [11], dental pulp [12], periodontal ligament [13], tendon [14], skin [15], muscle [16], and UC [17].

BM is considered the traditional source of MSCs, and the characteristics and application of BM-MSCs have been widely studied. However, BM isolation is an invasive and painful procedure that may cause hemorrhage, infection, and, in some cases, chronic pain [18]. Furthermore, BM-MSCs have been reported to show accelerated senescence following the donors age [19]. Also harvesting adipose in order to isolate AD-MSCs needs surgery and it accompanies invasiveness to some extent. Harvesting UCB doesn’t need surgery, while UCB is harvested only when it is available with aseptic procedure, and UCB often cannot be collected cleanly depending on the condition of the delivery and the baby. On the other hand, the UC is often discarded as medical materials, therefore isolating UC is painless and noninvasive, and UC can always be collected unlike UCB. Furthermore UC-MSCs have multipotency properties comparable to those of MSCs derived from other tissues [13, 20]. Furthermore, UC-MSCs have attracted attention from their immunomodulatory properties. They express human leukocyte antigen (HLA)-class I less than BM-MSCs [21] and are thus less immunogenic. For the purpose of treatment in the acute phase of TBI, it is considered difficult to prepare the patient for autologous BM- or AD-MSCs transplantation. Therefore, we are focusing on the UC as a major source of MSCs with an important potential for cell therapy, as they are suitable for allogeneic transplantation.

There are several protocols for isolating and culturing UC-MSCs. The improved explant method is generally used [22]; UC is minced into small fragments, and then seeded regularly on a tissue culture dish. Culture media is renewed every 3 days until fibroblast-like adherent cells reach 80–90% confluency. After that, cells are detached using trypsin. These cells are called passage 1 UC-MSCs, and this passage 1 MSCs are mass-cultured to passage 4 MSCs, which are usually destined for basic and clinical use.

For the treatment of CNS injury and sequelae, cryopreservation of MSCs is necessary, because cell therapy should be performed at any given time, either in the acute phase of injury or the chronic phase, when rehabilitation is attempted. Therefore, considering cell viability, cell therapeutics should generally be cryopreserved and thawed just before use. Moreover, long-term cryopreservation of MSCs is necessary, potentially because the same donor sample may be required for multiple times; thus this procedure should be further investigated in the future. Cryoprotectants are used to prevent damage in slow freezing [23]. Recently, we demonstrated the cryopreservation of UC tissue using a serum- and xeno origin-free cryoprotectant, STEM-CELLBANKER® (Zenoaq, Fukushima, Japan), and demonstrated that UC-MSCs cryopreserved retained phenotypes characteristic of MSCs, including immunosuppressive activity [24, 25]. These MSCs cryopreserved in a tube can be used as cell therapeutics for patients in any phase of the traumatic CNS injury.

3. Characteristics of MSCs

The minimal criteria for defining MSCs are following: First, MSCs must be adherent cells when maintained in standard culture conditions. Second, MSCs need to express surface markers of MSCs: CD105, CD73, and CD90, but not CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR. Third, MSCs must differentiate into adipocytes, chondroblasts, and osteoblasts in vitro [26, 27]. These criteria are common to all sources of MSCs.

The major properties of MSCs useful in the treatment of traumatic CNS injury are immunomodulation and neurotropism because most of traumatic CNS injury accompanies inflammation to some extent and neuronal damage. By the virtue of these properties, suppression of inflammation, neuroprotection and neurorestoration are enabled in the injured area of the CNS.

3.1 Immunomodulatory properties

Immunomodulatory effects are the most popular property of MSCs for clinical use [28, 29]. The lack of HLA-class II in MSCs lead to prevention from recognition by CD4 positive helper T-cells [30]. In addition, MSCs can inhibit the proliferation of immune cells and their cytokine production [31, 32, 33, 34]. The immunomodulation may be the result of MSCs releasing factors such as indoleamine 2,3-dioxygenase (IDO), prostaglandin E (PGE2), HLA-G5 modulating the functions of T cells [35], hepatocyte growth factor (HGF), and tumor growth factor beta 1 (TGF-β1) (Fig. 1) [36]. The immunosuppressive potential of MSCs is induced in the presence of inflammatory cytokines such as gamma interferon (IFNγ) and tumor necrosis factor alpha (TNFα) [37]. In contrast, with low levels of inflammatory cytokines, MSCs enhance T-cell activation and act as inducers of inflammation [38]. We also demonstrated that UC-MSCs cause inflammation with elevated expression of inflammatory cytokine such as interleukin 1 beta (IL-1β) in resting microglia in some lots [39]. These abilities of immunomodulation are also regulated via Toll-like receptors (TLRs) [40, 41]. As for the difference between sources of MSCs in immunomodulatory properties, only a few studies directly compared the immunomodulatory potency of MSCs from different tissue sources, and the results revealed there is no significance in the immunomodulatory ability of MSCs from different sources [20]. Overall, this immunomodulatory property, regardless of the source of MSC, is thought to play a key role in the therapeutic effect of MSCs in traumatic CNS injuries.

Fig. 1.

Characteristics of MSCs and application for traumatic CNS injury. MSCs derived from various tissues secret several neurotrophic factors and immunomodulatory factors. These abilities of tissue repair and immunomodulation are expected as cell therapeutics for the treatment of traumatic CNS injury.

3.2 Neurotrophic properties

MSCs have been reported to secrete heterogeneous lipid vesicles called extracellular vesicles (EVs), which act as mediators for inter-cell communication [42, 43]. These EVs including exosomes secreted by MSCs are known to improve neuronal functions in models of neurological injury [43, 44]. We have also demonstrated the amelioration of neuronal injury followed by functional improvement in mice models after MSC administration, which was the result of trophic factors secretion rather than neuronal differentiation and eternal cell replacement by MSCs [45]. We also showed in an in vitro experiment that brain-derived neurotrophic factor (BDNF) and HGF secreted by UC-MSCs improved neuronal injury, as indicated by an increase in the immature neuron, neurite outgrowth, and cell proliferation, and also a reduction the number of neurons with apoptosis/necrosis [46]. These properties of neurogenesis and neuroprotection in MSCs could be the major mechanisms in treating traumatic CNS injuries. Furthermore, genetically modified MSCs are studied for treatment of traumatic CNS injury. The efficacy of genetically engineered MSC strongly expressing neurotrophic factors has been reported in TBI models [47].

4. Effects of MSCs in traumatic neurological injury models

The neuroinflammation accompanying the increase in reactive astrocytes and the activation of microglia following traumatic CNS injury has been reported as an important mechanism [48, 49]. The trauma causes a multifaceted pathophysiological processes leading to the glial scar [49]. Increased reactive astrocytes are commonly observed in TBI. Astrocytes, through multiple bioactive factors, exert beneficial roles in TBI, including promotion and restriction of neurogenesis and synaptogenesis, modulation of neuroinflammation, and disruption and repair of the blood brain barrier (BBB) [50, 51]. MSCs reportedly have the ability to reduce this reactive gliosis, leading to neurological amelioration [45, 52]. Microglia are immune responder in the CNS that can switch to an activated microglia (M1) henotype secreting proinflammatory cytokines; conversely, they can turn to a resting microglia (M2) phenotype secreting anti-inflammatory cytokines and neurotrophic factors [53]. It is reported that after TBI, M2-microglia exert anti-inflammatory effects by releasing anti-inflammatory cytokines, resulting in tissue repair and regeneration facilitating phagocytosis [48]. Therefore, microglial polarization could be a target in treatment for TBI. We demonstrated that UC-MSCs exert immunomodulatory effects and change the phenotype of activated microglia to resting microglia, by suppressing the expression of IL-1β and phospho nuclear factor-kappa B (pNFκB) which plays a key role in inflammatory pathway in activated microglia, and changing their morphology from ameboid to ramified; this process is achieved by increasing active Rho guanosine triphosphatase (GTPase) through the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway which control the GTPase expression[39].

In the basic research using MSCs for traumatic CNS injury animal models, many reports have demonstrated the efficacy of MSCs and their secretomes (exosomes) for neurological improvement in traumatic CNS injury models. Kumar Mishra et al. [54] found an improvement in tissue, as well as functional behaviors in MSC-infused TBI mice. AD-MSCs-derived exosomes are reported to promote functional recovery, suppress neuroinflammation, reduce neuronal apoptosis, and increase neurogenesis in rats with TBI; this is achieved by suppressing microglia activation [55]. In addition, Xu et al. [47] reported that MSCs-derived exosomes promote a BDNF-mediated neurogenesis and inhibit apoptosis in rats with TBI. Exosomes from MSCs have also shown potential in healing SCI in a rat model through inhibition of pericyte migration, which improved motor functioning and the structural integrity of the blood-spinal cord barrier [56]. Li et al. [57] demonstrated the inhibition of neuronal apoptosis via activation of the Wnt/beta-catenin signaling pathway by MSCs in an SCI model. The comparison of therapeutic effects of AD-MSCs, BM-MSCs, and cranial bone-derived MSCs on chronic SCI model rats have been reported [58]. As compared with AD- and BM-MSCs, cranial bone-derived MSCs highly expressed many neurotrophic factors which improved motor function in chronic SCI model. On the other hand, comparison of efficacy in BM-MSCs and UC-MSCs transplantations for SCI model showed that both MSCs reduced neuropathic pain and resulted in subsequent motor recovery after SCI, while survival rate and electrophysiological findings of UC-MSCs were significantly better than BM-MSCs [59]. These basic research data enable us to develop a protocol of clinical trials using MSCs for traumatic CNS injuries.

5. Clinical studies using MSCs for traumatic CNS injuries

Based on the mechanisms and efficacy suggested by the basic research mentioned above, many clinical trials using MSCs for traumatic CNS injury have been conducted [60]. Recent clinical trials using MSCs for traumatic CNS injuries are summarized in Table 1 (Ref. [60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72]). Regarding the origin of MSCs, BM, UC, UCB, and AD have all been widely used as sources. As for the administration route of MSCs, in most clinical studies, MSCs are administered by intrathecal injection (IT) or direct infusion into the injured sites. These clinical trials have mainly reported on the feasibility and efficacy of MSC therapies for neurological disorders, with no severe adverse events.

Table 1.Recent clinical trials using MSCs for traumatic CNS injuries (modified reference) [60].
Reference Traumatic CNS injuries Source of MSCs Number of patients Mean age, year Administration route Cell dose Number of administration Results Adverse events
Experimental Contro1
Wang et al., 2013 [61] TBI UC 20 20 27.5 ± 9.4 IT 6.0 × 107 cells 4 Comprehensive functional recovery and improvement in the ADL after 6 months Mild dizziness, headache
28.6 ± 10.1
Tian et al., 2013 [62] TBI BM 97 0 21.1 IT 3.0–5.0 × 106 cells 1 Improvement of consciousness and motor function after 14 days No
35.3
Xiao et al., 2018 [63] SCI UCB 2 0 28 Transplantation into the lesion with collagen scaffolds 4 × 107 cells 1 Improvement of motor function after 3, 6, 12 months No
30
Improvement of sensory function after 2, 4, 12 months
Vaquero et al., 2019 [64] SCI BM 11 0 44.91 IT 3 × 100 × 106 cells 3 Motor, sensory and bladder-bowel functional improvement after 4, 7, 10 months No
(28–62)
Vaquero et al., 2017 [65] SCI BM 10 0 42.2 IT 30 × 106 cells at 3-months interval 4 Motor, sensory and bladder-bowel functional improvement after 3, 6, 9, 12 months Head ache
Satti et al., 2016 [66] SCI BM 9 0 31.6 IT 1.2 × 106 /kg at 4 weeks interval 2/3 Safety assessment only No
(24–38)
Oh et al., 2016 [67] SCI BM 16 0 40.9 Direct injection into the lesion + IT 1.6 × 107 cells 1 Very weak therapeutic efficacy after 6 months Sensory deterioration, muscle rigidity, tingling sense
(18–65) 3.2 × 107 cells
Hur et al., 2016 [68] SCI AD 14 0 41.9 IT 3 × 3 × 107 3 Improvement of function after 8 months Nausea, head ache, vomit
Mendonça et al., 2014 [69] SCI BM 14 0 35.7 Direct injection into the lesion 5 × 106 cells/cm3 per lesion volume 1 Motor, sensory and bladder-bowel functional improvement after 6 months Pain, cerebrospinal fluid leak
(23–61)
Cheng et al., 2014 [70] SCI UC 10 34 35.3 Direct injection into the lesion 2 × 2 × 107 cells 2 Motor, sensory and bladder functional improvement after 6 months Radiating neuralgia
(19–57)
Superior efficacy than that of rehabilitation therapy
Dai et al., 2013 [71] SCI BM 20 20 22–54 Direct injection into the lesion 20 × 106 cells 1 Improvement of motor, sensory and bladder function after 6 months Fever, headache, pain
Karamouzian et al., 2012 [72] SCI BM 11 20 33.2 IT 0.7–1.2 × 106 cells 1 Possible efficacy in the motor and sensory function No
(23–48)

As for TBI, Wang et al. [61] demonstrated the results of a phase 2 clinical trial using UC-MSCs for patients with sequelae of TBI. In this study, UC-MSCs administration improved the patients’ neurological function and self-care ability after 6 months. Moreover, Tian et al. [62] reported the clinical therapeutic effects and safety of autologous BM-MSCs therapy for TBI. The results showed improvement in the brain function, post-therapeutic improvements in consciousness, and motor functions. Additionally, they showed that the patients’ age and the time between the injury and therapy influenced the outcomes of the cellular therapy; however, no correlation was found between the number of cell injections and therapeutic improvements. This last result is of great importance in deciding the protocol of administration.

As for the clinical trials involving MSC treatment for SCI, Vaquero et al. [64] reported that patients who were administered BM-MSCs showed clinical improvements in sensitivity, motor power, spasticity, neuropathic pain, sexual function, and/or sphincter dysfunction, regardless of the level/degree of injury, age, or time elapsed since the SCI. They also showed the efficacy and feasibility of administration of repeated doses of BM-MSCs [65]. Ten patients with incomplete SCI received administrations of autologous BM-MSCs at 1, 4, 7 and 10 months; all of them showed some extent of improvement in sensitivity and motor function. Interestingly, after three administrations of MSCs, the mean values of BDNF, glial-derived neurotrophic factor, ciliary neurotrophic factor, and neurotrophin 3/4 slightly increased compared with basal levels. Hur et al. [68] showed the effects and feasibility of autologous AD-MSCs transplantation in patients with SCI. Over the 8-month follow-up, the patients who received AD-MSCs did not experience any serious adverse events, and several patients showed mild improvements in neurological function. Transplanting collagen scaffolds with human UC-MSCs has also been reported to have therapeutic potential as a treatment for SCI. Collagen scaffolds with human UC-MSCs were transplanted directly into the injury site, and the recovery of sensory and motor functions was observed in both patients [66]. Oh et al. [67] reported on the injection of autologous BM-MSCs into the intramedullary area and subdural space, and concluded that this single MSCs application was safe, despite having a very weak therapeutic effect compared to multiple MSC injections. These clinical trials reporting the efficacy and feasibility of MSCs encourage us to conduct further large-scale clinical studies using MSCs for traumatic CNS injury.

6. Challenges and perspectives

In clinical studies, many sources of MSCs are used, which makes it difficult to have a consensus about the appropriate protocol for the treatment of traumatic CNS injuries. Additionally, variations in culture media, isolation methods, cell counts methods, the number of passages, donor age, delivery methods and finally host receptibility might further blur the potential differences among MSCs therapeutic effects. MSCs feature numerous advantages and one of them is low tumorigenesis risks [73, 74]. However, the possibility of tumorigenesis in MSCs are always discussed. MSCs have been reported to possess the properties of both tumor suppression and promotion [75, 76, 77]. On the other hand, EVs including exosome secreted from MSCs have no risk of tumorigenesis. Genetically engineered MSCs are also attracting attention for treatment of traumatic CNS injury. By using genetically engineered MSC, which strongly expresses neurotrophic factors, are reported to be effective for TBI at the basic experimental stage [47]. These genetically modified MSCs also have the potential to expand the possibilities of treating traumatic CNS injury. These secretome of MSCs and also genetically engineered MSCs might be future possible therapeutics for treatment of traumatic CNS injuries. Furthermore, since MSCs are used to treat skin injuries with wound healing [78], MSCs may also be effective in skin and soft tissue trauma in traumatic CNS injury. In the future, all injured sites might be treated by MSCs in patients with traumatic CNS injury.

7. Conclusions

Recent clinical trials indicate that the use of MSCs as a new cell therapy is expected to be effective in combination with conventional rehabilitation and other medication.

Regarding allogeneity, autologous transplantation might be desirable when considering the possibility of rejection of the host, but this completely depends on the MSCs source. Isolation of autologous BM- or AD-MSCs in adults with traumatic CNS injury is possible, while autologous UC- or UCB-MSCs in adults is very difficult because this would have required cryopreservation of these cells decades ago. Although it is related to the timing of preparation and administration of MSCs. Previous studies have mainly used MSCs for the purpose of treating sequelae and recovering function after the acute phase. In anticipation of the immunomodulatory effect, administration of MSCs immediately after injury should also be considered.

As for the administration route, many clinical trials have opted for IT or direct infusion into injured sites because of the existence of the BBB, which would make it difficult for the intravenously injected MSCs to migrate to the injured CNS sites. In our study, most of the intravenously injected UC-MSCs were trapped in the lungs and could not reach the injury site in the brain [45].

Traumatic CNS injuries have multiple causes and their pathogenesis involves multiple factors, therefore cell therapeutics that have immunomodulatory functions and secrete neurotrophic factors might be more suitable candidates for CNS injury therapeutics.

The prognosis of patients with traumatic CNS injury remains abysmal, with a high mortality rate. It would be necessary to further investigate the appropriate protocol for MSC administration, and large-scale clinical studies on using MSCs to treat traumatic CNS injury will extend the possibility of MSCs therapy in the future.

Abbreviations

CNS, central nervous system; TBI, traumatic brain injury; SCI, spinal cord injury; MSCs, mesenchymal stromal cells; UC, umbilical cord; UC-MSCs, umbilical cord-derived mesenchymal stromal cells; BM, bone marrow; UCB, umbilical cord blood; AD, adipose tissue; IDO, indoleamine 2,3-dioxygenase; PGE2, prostaglandin E2; TGF-β1, tumor growth factor beta 1; IFNγ, gamma interferon; TNFα, tumor necrosis factor alpha; TLR, Toll-like receptor; HLA, human leukocyte antigen; IL-1β, interleukin 1 beta; pNFκB, phospho nuclear factor-kappa B; GTPase, guanosine triphosphatase; Akt, protein kinase B; HGF, hepatocyte growth factor; EV, extracellular vesicle; BDNF, brain-derived neurotrophic factor; BBB, blood brain barrier; PI3K, phosphoinositide 3-kinase; IT, intrathecal injection.

Author contributions

Conception and design of the study—TM, KS and TNI. Acquisition of data—TM and KS. Drafting or revising the manuscript—All authors. All authors have approved the final article.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

The authors thank all members of the Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo for their help and consultation during this work.

Funding

This study was supported by Japan Society for the Promotion of Science (JSPS KAKENHI Grant Number 20K22892 and 21K15618).

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Xu L, Ye X, Wang Q, Xu B, Zhong J, Chen Y, et al. T-cell infiltration, contribution and regulation in the central nervous system post-traumatic injury. Cell Proliferation. 2021; 54: e13092.
[2]
Michinaga S, Koyama Y. Pathophysiological Responses and Roles of Astrocytes in Traumatic Brain Injury. International Journal of Molecular Sciences. 2021; 22: 6418.
[3]
Lin J, Xiong Z, Gu J, Sun Z, Jiang S, Fan D, et al. Sirtuins: Potential Therapeutic Targets for Defense against Oxidative Stress in Spinal Cord Injury. Oxidative Medicine and Cellular Longevity. 2021; 2021: 1–14.
[4]
Fleminger S, Oliver DL, Lovestone S, Rabe-Hesketh S, Giora A. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. Journal of Neurology, Neurosurgery and Psychiatry. 2003; 74: 857–862.
[5]
Schmidt S, Kwee LC, Allen KD, Oddone EZ. Association of ALS with head injury, cigarette smoking and APOE genotypes. Journal of the Neurological Sciences. 2010; 291: 22–29.
[6]
Wirdefeldt K, Adami H, Cole P, Trichopoulos D, Mandel J. Epidemiology and etiology of Parkinson’s disease: a review of the evidence. European Journal of Epidemiology. 2011; 26: S1–S58.
[7]
Narayan RK, Michel ME, Ansell B, Baethmann A, Biegon A, Bracken MB, et al. Clinical trials in head injury. Journal of Neurotrauma. 2002; 19: 503–557.
[8]
Aertker BM, Bedi S, Cox CS. Strategies for CNS repair following TBI. Experimental Neurology. 2016; 275: 411–426.
[9]
Gnecchi M, Melo LG. Bone marrow-derived mesenchymal stem cells: isolation, expansion, characterization, viral transduction, and production of conditioned medium. Methods in Molecular Biology. 2009; 482: 281–294.
[10]
Bieback K, Klüter H. Mesenchymal stromal cells from umbilical cord blood. Current Stem Cell Research & Therapy. 2007; 2: 310–323.
[11]
Gruber HE, Deepe R, Hoelscher GL, Ingram JA, Norton HJ, Scannell B, et al. Human Adipose-Derived Mesenchymal Stem Cells: Direction to a Phenotype Sharing Similarities with the Disc, Gene Expression Profiling, and Coculture with Human Annulus Cells. Tissue Engineering Part A. 2010; 16: 2843–2860.
[12]
Chen TF, Chen KW, Chien Y, Lai YH, Hsieh ST, Ma HY, et al. Dental Pulp Stem Cell-Derived Factors Alleviate Subarachnoid Hemorrhage-Induced Neuroinflammation and Ischemic Neurological Deficits. International Journal of Molecular Sciences. 2019; 20: 3747.
[13]
Kim J, Jo CH, Kim H, Hwang Y. Comparison of Immunological Characteristics of Mesenchymal Stem Cells from the Periodontal Ligament, Umbilical Cord, and Adipose Tissue. Stem Cells International. 2018; 2018: 8429042.
[14]
Liu C, Luo J, Zhang K, Lin L, Liang T, Luo Z, et al. Tendon-Derived Stem Cell Differentiation in the Degenerative Tendon Microenvironment. Stem Cells International. 2018; 2018: 2613821.
[15]
Orciani M, Di Primio R. Skin-derived mesenchymal stem cells: isolation, culture, and characterization. Methods in Molecular Biology. 2013; 989: 275–283.
[16]
Camernik K, Mihelic A, Mihalic R, Marolt Presen D, Janez A, Trebse R, et al. Skeletal-muscle-derived mesenchymal stem/stromal cells from patients with osteoarthritis show superior biological properties compared to bone-derived cells. Stem Cell Research. 2019; 38: 101465.
[17]
Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells. 2003; 21: 105–110.
[18]
Nagamura-Inoue T, Mukai T. Umbilical Cord is a Rich Source of Mesenchymal Stromal Cells for Cell Therapy. Current Stem Cell Research & Therapy. 2017; 11: 634–642.
[19]
Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2004; 33: 919–926.
[20]
Mattar P, Bieback K. Comparing the Immunomodulatory Properties of Bone Marrow, Adipose Tissue, and Birth-Associated Tissue Mesenchymal Stromal Cells. Frontiers in Immunology. 2015; 6: 560.
[21]
Lu L, Liu Y, Yang S, Zhao Q, Wang X, Gong W, et al. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials. Haematologica. 2006; 91: 1017–1026.
[22]
Mori Y, Ohshimo J, Shimazu T, He H, Takahashi A, Yamamoto Y, et al. Improved explant method to isolate umbilical cord-derived mesenchymal stem cells and their immunosuppressive properties. Tissue Engineering. Part C, Methods. 2015; 21: 367–372.
[23]
Meryman HT. Cryoprotective agents. Cryobiology. 1971; 8: 173–183.
[24]
Shimazu T, Mori Y, Takahashi A, Tsunoda H, Tojo A, Nagamura-Inoue T. Serum- and xeno-free cryopreservation of human umbilical cord tissue as mesenchymal stromal cell source. Cytotherapy. 2016; 17: 593–600.
[25]
Shimazu T, Mori Y, Takahashi A, Tsunoda H, Tojo A, Nagamura-Inoue T. Serum- and xeno-free cryopreservation of human umbilical cord tissue as mesenchymal stromal cell source. Cytotherapy. 2016; 17: 593–600.
[26]
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. the International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8: 315–317.
[27]
Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al. Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy. 2006; 7: 393–395.
[28]
Nagamura-Inoue T. Umbilical cord-derived mesenchymal stem cells: their advantages and potential clinical utility. World Journal of Stem Cells. 2014; 6: 195-202.
[29]
Wang Y, Chen X, Cao W, Shi Y. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nature Immunology. 2015; 15: 1009–1016.
[30]
Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 2003; 101: 3722–3729.
[31]
English K, Barry FP, Mahon BP. Murine mesenchymal stem cells suppress dendritic cell migration, maturation and antigen presentation. Immunology Letters. 2008; 115: 50–58.
[32]
Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008; 111: 1327–1333.
[33]
Asari S, Itakura S, Ferreri K, Liu C, Kuroda Y, Kandeel F, et al. Mesenchymal stem cells suppress B-cell terminal differentiation. Experimental Hematology. 2009; 37: 604–615.
[34]
Magatti M, De Munari S, Vertua E, Gibelli L, Wengler GS, Parolini O. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells. 2008; 26: 182–192.
[35]
Seshareddy K, Troyer D, Weiss ML. Method to isolate mesenchymal-like cells from Wharton’s Jelly of umbilical cord. Methods in Cell Biology. 2008; 86: 101–119.
[36]
Zhou C, Yang B, Tian Y, Jiao H, Zheng W, Wang J, et al. Immunomodulatory effect of human umbilical cord Wharton’s jelly-derived mesenchymal stem cells on lymphocytes. Cellular Immunology. 2011; 272: 33–38.
[37]
Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008; 2: 141–150.
[38]
Li W, Ren G, Huang Y, Su J, Han Y, Li J, et al. Mesenchymal stem cells: a double-edged sword in regulating immune responses. Cell Death and Differentiation. 2013; 19: 1505–1513.
[39]
Mukai T, Di Martino E, Tsuji S, Blomgren K, Nagamura-Inoue T, Ådén U. Umbilical cord-derived mesenchymal stromal cells immunomodulate and restore actin dynamics and phagocytosis of LPS-activated microglia via PI3K/Akt/Rho GTPase pathway. Cell Death Discovery. 2021; 7: 46.
[40]
Raicevic G, Rouas R, Najar M, Stordeur P, Id Boufker H, Bron D, et al. Inflammation modifies the pattern and the function of Toll-like receptors expressed by human mesenchymal stromal cells. Human Immunology. 2010; 71: 235–244.
[41]
Waterman RS, Tomchuck SL, Henkle SL, Betancourt AM. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS ONE. 2010; 5: e10088.
[42]
Fuloria S, Subramaniyan V, Dahiya R, Dahiya S, Sudhakar K, Kumari U, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Regenerative Potential and Challenges. Biology-Basel. 2021; 10: 172.
[43]
Park KS, Bandeira E, Shelke GV, Lässer C, Lötvall J. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Research & Therapy. 2019; 10: 288.
[44]
Nakano M, Fujimiya M. Potential effects of mesenchymal stem cell derived extracellular vesicles and exosomal miRNAs in neurological disorders. Neural Regeneration Research. 2021; 16: 2359–2366.
[45]
Mukai T, Mori Y, Shimazu T, Takahashi A, Tsunoda H, Yamaguchi S, et al. Intravenous injection of umbilical cord-derived mesenchymal stromal cells attenuates reactive gliosis and hypomyelination in a neonatal intraventricular hemorrhage model. Neuroscience. 2018; 355: 175–187.
[46]
Mukai T, Tojo A, Nagamura-Inoue T. Umbilical Cord-Derived Mesenchymal Stromal Cells Contribute to Neuroprotection in Neonatal Cortical Neurons Damaged by Oxygen-Glucose Deprivation. Frontiers in Neurology. 2018; 9: 466.
[47]
Xu H, Jia Z, Ma K, Zhang J, Dai C, Yao Z, et al. Protective effect of BMSCs-derived exosomes mediated by BDNF on TBI via miR-216a-5p. Medical Science Monitor. 2020; 26: e920855.
[48]
Huang X, You W, Zhu Y, Xu K, Yang X, Wen L. Microglia: a Potential Drug Target for Traumatic Axonal Injury. Neural Plasticity. 2021; 2021: 1–13.
[49]
Tran AP, Warren PM, Silver J. New insights into glial scar formation after spinal cord injury. Cell and Tissue Research. 2021.
[50]
Castejón OJ. Morphological astrocytic changes in complicated human brain trauma. A light and electron microscopic study. Brain Injury. 1998; 12: 409–427.
[51]
Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive astrocytes protect tissue and preserve function after spinal cord injury. Journal of Neuroscience. 2004; 24: 2143–2155.
[52]
Kim S, Kim YE, Hong S, Kim K, Sung DK, Lee Y, et al. Reactive microglia and astrocytes in neonatal intraventricular hemorrhage model are blocked by mesenchymal stem cells. Glia. 2020; 68: 178–192.
[53]
Tsuji S, Di Martino E, Mukai T, Tsuji S, Murakami T, Harris RA, et al. Aggravated brain injury after neonatal hypoxic ischemia in microglia-depleted mice. Journal of Neuroinflammation. 2020; 17: 111.
[54]
Kumar Mishra S, Khushu S, Gangenahalli G. Neuroprotective response and efficacy of intravenous administration of mesenchymal stem cells in traumatic brain injury mice. European Journal of Neuroscience. 2021.
[55]
Chen Y, Li J, Ma B, Li N, Wang S, Sun Z, et al. MSC-derived exosomes promote recovery from traumatic brain injury via microglia/macrophages in rat. Aging. 2020; 12: 18274–18296.
[56]
Lu Y, Zhou Y, Zhang R, Wen L, Wu K, Li Y, et al. Bone Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Recovery Following Spinal Cord Injury via Improvement of the Integrity of the Blood-Spinal Cord Barrier. Frontiers in Neuroscience. 2019; 13: 209.
[57]
Li C, Jiao G, Wu W, Wang H, Ren S, Zhang L, et al. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/β-catenin Signaling Pathway. Cell Transplantation. 2019; 28: 1373–1383.
[58]
Otsuka T, Maeda Y, Kurose T, Nakagawa K, Mitsuhara T, Kawahara Y, et al. Comparisons of neurotrophic effects of mesenchymal stem cells derived from different tissue on chronic spinal cord injury rats. Stem Cells and Development. 2021.
[59]
Yousefifard M, Nasirinezhad F, Shardi Manaheji H, Janzadeh A, Hosseini M, Keshavarz M. Human bone marrow-derived and umbilical cord-derived mesenchymal stem cells for alleviating neuropathic pain in a spinal cord injury model. Stem Cell Research & Therapy. 2016; 7: 36.
[60]
Mukai T, Sei K, Nagamura-Inoue T. Mesenchymal Stromal Cells Perspective: New Potential Therapeutic for the Treatment of Neurological Diseases. Pharmaceutics. 2021; 13: 1159.
[61]
Wang S, Cheng H, Dai G, Wang X, Hua R, Liu X, et al. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Research. 2014; 1532: 76–84.
[62]
Tian C, Wang X, Wang X, Wang L, Wang X, Wu S, et al. Autologous bone marrow mesenchymal stem cell therapy in the subacute stage of traumatic brain injury by lumbar puncture. Experimental and Clinical Transplantation. 2013; 11: 176–181.
[63]
Xiao Z, Tang F, Zhao Y, Han G, Yin N, Li X, 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 Transplantation. 2019; 27: 907–915.
[64]
Vaquero J, Zurita M, Rico MA, Aguayo C, Bonilla C, Marin E, et al. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: Safety and efficacy of the 100/3 guideline. Cytotherapy. 2019; 20: 806–819.
[65]
Vaquero J, Zurita M, Rico MA, Bonilla C, Aguayo C, Fernández C, et al. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 2017; 19: 349–359.
[66]
Satti HS, Waheed A, Ahmed P, Ahmed K, Akram Z, Aziz T, et al. Autologous mesenchymal stromal cell transplantation for spinal cord injury: a Phase i pilot study. Cytotherapy. 2016; 18: 518–522.
[67]
Oh SK, Choi KH, Yoo JY, Kim DY, Kim SJ, Jeon SR. A Phase III Clinical Trial Showing Limited Efficacy of Autologous Mesenchymal Stem Cell Therapy for Spinal Cord Injury. Neurosurgery. 2016; 78: 436–447.
[68]
Hur JW, Cho T, Park D, Lee J, Park J, Chung Y. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: a human trial. Journal of Spinal Cord Medicine. 2018; 39: 655–664.
[69]
Mendonça MVP, Larocca TF, de Freitas Souza BS, Villarreal CF, Silva LFM, Matos AC, et al. Safety and neurological assessments after autologous transplantation of bone marrow mesenchymal stem cells in subjects with chronic spinal cord injury. Stem Cell Research & Therapy. 2015; 5: 126.
[70]
Cheng H, Liu X, Hua R, Dai G, Wang X, Gao J, et al. Clinical observation of umbilical cord mesenchymal stem cell transplantation in treatment for sequelae of thoracolumbar spinal cord injury. Journal of Translational Medicine. 2014; 12: 253.
[71]
Dai G, Liu X, Zhang Z, Yang Z, Dai Y, Xu R. Transplantation of autologous bone marrow mesenchymal stem cells in the treatment of complete and chronic cervical spinal cord injury. Brain Research. 2013; 1533: 73–79.
[72]
Karamouzian S, Nematollahi-Mahani SN, Nakhaee N, Eskandary H. Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clinical Neurology and Neurosurgery. 2012; 114: 935–939.
[73]
Mannino G, Russo C, Longo A, Anfuso CD, Lupo G, Lo Furno D, et al. Potential therapeutic applications of mesenchymal stem cells for the treatment of eye diseases. World Journal of Stem Cells. 2021; 13: 632–644.
[74]
Öner A. Stem Cell Treatment in Retinal Diseases: Recent Developments. Turkish Journal of Ophthalmology. 2019; 48: 33-38.
[75]
Timaner M, Tsai KK, Shaked Y. The multifaceted role of mesenchymal stem cells in cancer. Seminars in Cancer Biology. 2020; 60: 225–237.
[76]
Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Molecular Cancer. 2017; 16: 31.
[77]
Li C, Zhao H, Wang B. Mesenchymal stem/stromal cells: Developmental origin, tumorigenesis and translational cancer therapeutics. Translational Oncology. 2021; 14: 100948.
[78]
Riedl J, Popp C, Eide C, Ebens C, Tolar J. Mesenchymal stromal cells in wound healing applications: role of the secretome, targeted delivery and impact on recessive dystrophic epidermolysis bullosa treatment. Cytotherapy. 2021; 16: S1465–S3249.
Share
Back to top