Next Article in Journal
Protein Quality Control of NKCC2 in Bartter Syndrome and Blood Pressure Regulation
Previous Article in Journal
Correction: Zhao et al. Hypermethylation of UCHL1 Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing Degradation of Cortactin (CTTN). Cells 2020, 9, 559
Previous Article in Special Issue
The Translation of Nanomedicines in the Contexts of Spinal Cord Injury and Repair
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molars to Medicine: A Focused Review on the Pre-Clinical Investigation and Treatment of Secondary Degeneration following Spinal Cord Injury Using Dental Stem Cells

by
Sandra Jenkner
1,2,
Jillian Mary Clark
2,3,
Stan Gronthos
1,4 and
Ryan Louis O’Hare Doig
2,3,*
1
School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia
2
Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
3
Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia
4
Mesenchymal Stem Cell Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
*
Author to whom correspondence should be addressed.
Cells 2024, 13(10), 817; https://doi.org/10.3390/cells13100817
Submission received: 1 April 2024 / Revised: 1 May 2024 / Accepted: 7 May 2024 / Published: 10 May 2024

Abstract

:
Spinal cord injury (SCI) can result in the permanent loss of mobility, sensation, and autonomic function. Secondary degeneration after SCI both initiates and propagates a hostile microenvironment that is resistant to natural repair mechanisms. Consequently, exogenous stem cells have been investigated as a potential therapy for repairing and recovering damaged cells after SCI and other CNS disorders. This focused review highlights the contributions of mesenchymal (MSCs) and dental stem cells (DSCs) in attenuating various secondary injury sequelae through paracrine and cell-to-cell communication mechanisms following SCI and other types of neurotrauma. These mechanistic events include vascular dysfunction, oxidative stress, excitotoxicity, apoptosis and cell loss, neuroinflammation, and structural deficits. The review of studies that directly compare MSC and DSC capabilities also reveals the superior capabilities of DSC in reducing the effects of secondary injury and promoting a favorable microenvironment conducive to repair and regeneration. This review concludes with a discussion of the current limitations and proposes improvements in the future assessment of stem cell therapy through the reporting of the effects of DSC viability and DSC efficacy in attenuating secondary damage after SCI.

1. Introduction

Spinal cord injury (SCI) is a debilitating condition caused by damage or disease of the spinal cord. It can result in long-term or permanent loss of mobility, sensation, and/or autonomic function due to the impaired conduction of descending and ascending neurotransmission. SCI consequently leads to an increased risk of premature mortality and often generates severe comorbidities, including, but not limited to, chronic neuropathic pain, sexual dysfunction, bowel and bladder dysfunction, immunocompromise, and mental health and well-being disturbances. This causes serious physical, environmental, societal, and economic burdens for patients and their families [1].
It is estimated that 900,000 new cases of traumatic SCI occur annually on a global scale [2], with the lifetime economic costs to individuals with SCI estimated between USD 1.2–2.5 million [3]. Importantly, these numbers are expected to rise with improved care and the increased life expectancy of individuals living with a SCI. Despite this high incidence and a wealth of knowledge on the pathophysiology of SCI, there is currently no treatment to halt or reverse the neurological deficits within the spinal cord or prevent the etiology of comorbidities.

2. Pathophysiology of the Secondary Injury Cascade following Spinal Cord Injury

The pathophysiology of traumatic SCI impacts the neural structures within the spinal cord as well as each glial population in different ways and involves a complex and unique multicellular response. Similar to other central nervous system (CNS) trauma (e.g., traumatic brain injury (TBI), concussion, and stroke), SCI involves both primary and secondary injury mechanisms. Primary injury refers to the immediate mechanical impact or physical disruption to the neural tissue (reviewed in [4]) that occurs at the time of trauma. While the complete transection of the spinal cord is rare, hemi-sections, partial tearing, or contusion injuries are more common in the clinical setting. Neural tissue surrounding the primary injury area becomes vulnerable to degeneration known as secondary injury, which involves biochemical and cellular cascades of events that exacerbate and propagate the initial injury (Figure 1) and worsen functional outcomes [5]. Secondary events can be temporally divided into acute (<48 h), subacute (48 h to 14 days), intermediate (14 days to 6 months), and chronic (>6 months) phases. However, secondary injury often lasts throughout an individual’s lifetime, preserving a hostile environment that prevents the complete healing or regeneration of the injured area. Given the progressive nature of the secondary injury cascade, the multiple pathological and physiological mechanisms involved provide important therapeutic targets. The following sections provide background information on the biochemical events, cascades, and biological factors observed during secondary injury after SCI that are relevant to the therapeutic actions of stem cells discussed later in this review.

2.1. Vascular Events—Hemorrhage and Edema

Typically, mechanical impacts to the spinal cord result in immediate disruption and damage to the surrounding microvasculature of the blood–spinal cord barrier (BSCB). Numerous hemorrhages occur within the lesion immediately, with gray matter most affected due to the total cessation of blood perfusion, which can last more than 24 h [6]. Ischemia also arises as a result of vasospasms, endothelial cell swelling, and edema [7]. When compromised by vascular events, the spinal cord becomes highly susceptible to further damage above and below the injury site. However, the extent of these events is dependent on the type and severity of the injury [8]. Within white and gray matter, the leakage of plasma fluid into the meningeal compartments and extracellular space, as well as loss of ionic homeostasis, is observed [7,9,10], including disturbances in intracellular calcium (Ca2+) levels that elicit swelling of blood vessels, neurons, and glia [11]. Blood–spinal cord barrier integrity is disrupted within 5 min up to 28 days post-injury [9,12], evidenced by the dissociation of pericytes surrounding microvessels [7] permitting the indiscriminate passage of substances mediated by endothelin-1 and matrix metalloproteinases (MMPs) [13]. Ischemic damage with further cell death is maintained chronically after injury due to systemic hypotension and hypoxic tissue damage [14], in part mediated by the loss of pericyte function [15]. Unfortunately, the re-establishment of normal blood flow to the spinal cord causes reperfusion injury, exposing neural tissue to destructive biochemical factors and sustained influxes of immune cells and inflammatory cytokines in the acute to intermediate phases of injury, triggering further secondary degeneration.

2.2. Biochemical Events

2.2.1. Excitotoxicity

Glutamate excitotoxicity is a complex pathological mechanism caused by the excessive or prolonged activation of excitatory receptors via glutamate [16,17], the major excitatory transmitter of the mammalian CNS. After SCI, glutamate is released from damaged neural tissue at neurotoxic levels [18], and insufficient clearance by surviving astrocytes leads to the prolonged excitatory activation of glutaminergic ionotropic N-methyl-D-aspartate (NMDA) and a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Glutamate excitotoxicity contributes to large intracellular Ca2+ fluxes into neurons and glia [19], resulting in mitochondrial dysfunction, the activation of various enzymes, free radical and nitric oxide formation, and neural and oxidative stress [18,20]. Furthermore, the dysregulation of NMDA and AMPA receptors coupled with the loss of adenosine triphosphate (ATP)-dependent ionic gradient regulation increases the toxic accumulation of sodium and water within axons (for a review, see [21]). This ionic imbalance causes swelling that contributes to further mechanical damage.

2.2.2. Oxidative Stress

Oxidative stress is induced by the overproduction of reactive oxygen species (ROS), such as free oxygen radicals or nitric oxide by-products, which reach damaging levels in the first few hours of injury. The spinal cord is particularly prone to oxidative damage due to limited antioxidant defense capabilities, a large presence of polyunsaturated fatty acid chains sensitive to oxidation [13], and mitochondria shown to be up to 10 times more sensitive to oxidative damage than mitochondria in the brain [22]. SCI patients also exhibit a paucity of plasma antioxidants and, thus, insufficient oxidative balance for up to 12 months after injury [23]. Mitochondrial dysfunction has been demonstrated to be an initial source and target of oxidative stress, caused by excessive intracellular calcium influx, the inability to sequester this calcium, and resultant ROS production [24]. End products of oxidative stress, 4-HNE and 3-NT, cause further excessive damage to mitochondria, resulting in increased permeability, respiratory dysfunction, and mitochondrial death [25]. Increased nitric oxide content and protein oxidation, evidenced through protein carbonyl and 3-NT increases, occur within 24 h of injury [26]. Lipid peroxidation, the oxidative attack of the phospholipid bilayer of cell membranes, occurs later in the injury stage and spreads along the length of axons, causing the disruption of membrane transport, ionic gradient imbalances, oligodendrocyte damage, membrane lysis, and the death of previously unaffected cells [14,27]. Particularly prevalent in SCI, ROS are also formed within hypoxic environments as a result of ischemia from the by-products of hemoglobin breakdown and the phagocytosis of myelin debris by inflammatory cells, as well as during the reperfusion of ischemic tissue [13], with neutrophils and microglia implicated as the main producers of ROS within the damaged spinal cord [28].

2.2.3. Apoptosis and Cell Death

Secondary injury cascades in the acute to intermediate phases, particularly excitotoxicity and oxidative stress, culminate in the apoptotic death of neurons and glial cells. Apoptosis begins within hours of the injury, with 90% of neurons lost in the first 8 h within the lesion site [29]. Oligodendrocyte apoptosis can begin as early as 15 min post-injury [29], spreading to otherwise unaffected cells distant from the lesion site and leading to extended demyelination, impaired nerve conduction, and axonal degeneration [30]. The apoptosis of cells in the spinal cord is initiated through multiple pathways. Caspase pathways, the most critical initiators of apoptosis, including caspase-3, caspase-8, and caspase-9, are highly activated within 1 h to 1 day post-injury [30,31]. The Bcl2/Bax pathway, which regulates mitochondrial-mediated apoptosis, is also significantly dysregulated post-injury, with the anti-apoptotic Bcl2 protein being continuously downregulated up to 3 weeks post-injury, and the pro-apoptotic Bax protein elevated in a time-dependent manner [32], leading to a continued cycle of cell death. The various contributions of other forms of programed cell death to the pathophysiology of SCI beyond the scope of this review, such as ferroptosis, pyroptosis, and autophagy, have been reviewed in [33].

2.3. Inflammatory Events—Neuroinflammation and Immune Cell Influx

The breakdown of the BSCB exacerbates the neuroinflammatory response, orchestrated by residential glial activation and infiltrating leukocytes (neutrophils, monocytes, and lymphocytes). This response occurs both within the acute lesion and distally, and results in the influx of further infiltrating immune cells and the activation of microglia/macrophages. Most prevalently after SCI, macrophages and microglia become activated after exposure to pro-inflammatory mediators, such as interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), or lipopolysaccharide (LPS) [34], and exhibit a phenotype that results in pro-inflammatory cytokine secretion, phagocytosis, and collateral damage [35]. Microglia/macrophages and astrocytes secrete high levels of pro-inflammatory interleukin (IL)-12, IL-23, IL-1β, TNF-α, and IL-6, and low levels of anti-inflammatory IL-10, IL-4, and IL-13 [36,37,38], which cause further damage to vulnerable neural tissue. An excessive and dysfunctional increase in pro-inflammatory cytokines, interferons, and prostaglandins in acute SCI contributes to a cyclical influx of inflammatory cells and injury exacerbation at later injury stages.
Influxes of ROS and protease releasing neutrophils, which appear within the first hour post-injury, peak at 24 h and begin to diminish by 48 h [28], promoting increased vascular permeability and leukocyte influx. B and T lymphocytes infiltrate the cord within the first few hours of injury, declining by 7 days [39]. Microglial macrophages exhibit the greatest activation at 3 and 7 days, whilst monocyte-derived macrophage infiltration is the greatest by 7 days and persists for weeks, months, or indefinitely [28,40]. Reactive astrocytes are most prominent at chronic time points, but begin to display aberrant hypertrophy and proliferation sub-acutely in the first stage of glial scar formation [41]. Interestingly, the inflammatory response within the spinal cord greatly exceeds that of similar traumatic injuries in the brain [42].

2.4. Structural Events—Axonal and Myelin Changes, Glial Scarring, and Wallerian Degeneration

Structural damage, including the dysfunction and degeneration of neuronal cell bodies, axons, and glia, contributes substantially to the loss of neurological function and poor prognoses following SCI. Significant axonal changes begin within 15 min of injury and evolve over the acute phase. Acute changes include axonal fragmentation, swelling, organelle spillage into the extracellular space, myelin sheath thinning, and calcification [43,44], resulting in the necrotic death of cells and tissue. Cells with greater damage experience accelerated necrotic death, and overall lesion severity dictates the number of lost neurons and glia [45]. The intermediate and chronic phase of injury is marked by the stabilization and maturation of the lesion. This involves the formation of permanent cystic cavities within the lesion epicenter filled with extracellular fluid, connective tissue, and macrophages. The lesion is subsequently surrounded by a glial scar barrier composed of pericytes, fibroblasts [46], reactive astrocytes, microglia, and their secreted products, including chondroitin sulfate proteoglycans (CSPGs) and NG2 proteoglycan [47].
Degenerating oligodendrocytes and myelin sheaths release factors, including neurite outgrowth inhibitor A (Nogo-A) [48] and myelin-associated glycoprotein, which activate the Rho-associated protein kinase (ROCK) pathway to initiate growth cone collapse and neurite retraction [49]. Additionally, oligodendrocyte precursor cell depletion contributes to myelination defects or the insufficient remyelination of damaged axons [50]. Myelin proteins from damaged myelin sheaths accumulate, causing auto-immune reactivity mediated by lymphocytes that causes further myelin destruction and tissue dysfunction [51]. Furthermore, damaged axons undergo the slow process of Wallerian degeneration, resulting in subsequent remote tissue damage [52]. The resultant tissue environment is both inhospitable and inhibitory to repair, remyelination, or de novo pathway development. Syrinx formation, which occurs in approximately 30% of patients, leads to the development of large, fluid-filled, and high-pressure cavities with no clear etiology, which can greatly extend the lesion. An ascending lesion can contribute to neurological deterioration, brainstem involvement, or late-onset neuropathic pain [53].

3. The Benefits of Stem Cells for SCI Therapy

Numerous therapeutics designed to counter secondary injury pathophysiology or promote spinal cord repair have been studied in various pre-clinical animal models, most commonly in rodents (reviewed in [54]). The most common therapies are pharmacological (drugs or trophic factors) and cell or cell-derived in nature [54,55]. Arguably, the most studied yet controversial pharmacological agent for the management of SCI in the clinical setting is methylprednisolone sodium succinate [56]—a synthetic glucocorticoid receptor agonist that reduces oxidative stress, excitotoxicity, and pro-inflammatory neuronal phagocytosis [57]. Whilst promising results have been demonstrated in pre-clinical studies, the results of recent pharmaceutical clinical trials lack significant outcomes [58]. As reviewed by Zhang et al., 2021, there is still no safe and effective pharmacological or non-pharmacological treatment for the restoration of neurobiological function [59]. The pathophysiology of SCI is mutifaceted and rich with therapeutic targets. However, most of the pre-clinical investigations have been focused on a single component of the complex injury cascade. A multifactorial or multimodal approach to secondary injury may be necessary in order to achieve realistic functional improvements. Additionally, due to the limited growth capacity of the CNS and prohibitive cellular–molecular environment, treatments must be able to create a microenvironment that is conducive to endogenous or exogenous repair, as well as contribute to the inter-dependent biological processes of cellular regeneration and tissue restoration [27].
The growing interest in the treatment of SCI using stem cells is attributed to their ability to replenish lost neural and glial cells, as well as foster an environment conducive to endogenous or exogenous repair. Niches harboring tancyte-like cells expressing neural precursor markers within the spinal cord ependymal region surrounding the central canal have been postulated as a potential source of reparative endogenous stem cells [60,61]. However, evidence for the functional complexity of this ependymal niche mainly derives from investigations of mice or rat neonates [62]. Contradictory evidence comes from more recent studies indicating important interspecies differences in spinal cord and stem cell niche anatomy. For example, in comparison to non-primate mammals, the human spinal cord lacks central canal patency and proliferative and regenerative potential of surrounding ependymal cells [63,64]. These findings support the use of exogenous stem cell transplantation as an SCI therapy. Numerous candidate stem cell subsets harvested from different tissue sources are under active evaluation at the pre-clinical and clinical trial phases of translation, including progenitor cells, induced pluripotent stem cells, and glial cells (for an extensive review, see [65]). However, for stem cell transplantation to navigate the translational pathway and be adopted as a clinical therapy, the cell type must be: either available from a stem cell biobank, or be readily and noninvasively available from a viable donor; rapidly and easily expandable with limited ethical considerations; and have an acceptable risk profile.

3.1. Mesenchymal Stem Cells

Mesenchymal stem cells (MSCs) are one of the most commonly utilized therapeutic stem cell subsets, demonstrating safety and efficacy in clinical trials [66]. These adult multipotent cells are derived from multiple tissue types, including bone marrow and adipose tissue, and characterized by their expression of CD105, CD73, and CD90 and their lack of the expression of hematopoietic markers, including CD45, CD34, CD19, and CD11b [67,68]. MSCs have demonstrated a capacity to differentiate into chondrocytes [69], myofibers [70], and osteoblasts [71]. The differentiation potential of MSCs into neuronal-like cells has previously been demonstrated [72,73] but equally challenged in recent decades [74]. MSCs offer several advantages over engineered stem cell subsets, including simple extraction, minimal ethical considerations, no reprogramming costs, and less epigenetic uncertainty [75,76]. Their limited tissue-specific differentiation capacity, low tumorgenicity [77], immunomodulatory capabilities, and low immunogenicity [78,79] make them a favorable treatment option for CNS injuries [77,80].

3.2. Dental Stem Cells

Dental stem cells (DSCs) are a group of MSC-like cells (Figure 2) [81] that are gaining traction for their application as a potential neuroprotective and neuro-regenerative tool. First isolated by Gronthos et al. in 2000 [82], DSC populations reside in specialized tissue [83], express MSC markers (including CD90, CD73, CD105, CD44, and STRO-1 [84]), and display multi-differentiation potential, with the capacity to give rise to osteo/odontogenic, adipogenic, and neurogenic cell lineages [85,86].
DSCs exhibit qualities that make them advantageous to the more intensively investigated MSC neuro-regenerative therapies (Figure 2). DSCs originate from the neural crest and express neural markers even before neural differentiation, including microtubule-associated protein-2 (MAP-2), glial fibrillary acidic protein (GFAP), nestin, and β-III tubulin [87], particularly compared to MSC expression profiles [88], which have the potential to aid in neural differentiation [89]. These cells exist at a higher density in dental tissues than stem cells within bone marrow niches, have a higher proliferation rate than other MSC populations including bone marrow MSCs (BMSCs) [82], and have demonstrated encouraging regenerative potential in both peripheral and neural tissue, often with enhanced capabilities for neural regeneration compared to MSCs [90]. Of importance to attempts to design viable SCI therapeutics, DSCs are extracted non-invasively, either from discarded dental tissue or molar tooth removal [91]. The simplicity of donor harvest greatly limits the ethical considerations involved with extraction, which present for alternative stem cell populations [75]. A significant advantage involves autologous engraftment, and aligned to this, a reduced risk of immune reactivity. Furthermore, DSCs lack the expression of the major histocompatibility complex class II receptor (MHC II), preventing antigen recognition by the immune system [92]. As such, xenotransplantation of human DSCs has been investigated in rodent models of SCI with no immunosuppressive regimens [93,94], demonstrating no toxicity or cell rejection. Therefore, the immunomodulatory capacity and low immunogenicity of DSCs also permits allogeneic engraftment without the use of immunosuppressants [80], important in acute cases of neurotrauma for SCI individuals with already immunocompromised immune systems.

4. DSC Modulation of Secondary Cascades after SCI

Mesenchymal and dental stem cells have been extensively investigated in the field of neuro-regeneration and have demonstrated the ability to modulate the lesion environment after SCI to attenuate secondary injury (Figure 3). Following a PubMed search using the search term strategies shown in Supplementary Table S1, the literature to date presents 30 pre-clinical xenogeneic in vivo animal studies of SCI reporting the therapeutic effects of DSCs alone, DSC-conditioned media (CM), DSC exosomes, or DSCs combined with scaffolds, hydrogels, or drug therapy. Most studies delivered an acute intraspinal dose of DSCs (1 × 105–2 × 106) in rat models of spinal compression or contusion. Almost all the literature reported the neuro-recovery benefits of DSC therapy; however, only nine (30%) studies reported on the effects of DSC therapy on three or more of the key secondary injury mechanisms described above. Structural and anti-inflammatory or structural and anti-apoptotic or cell death events were mostly reported, whereas the reporting of the attenuation of biochemical events, such as glutamate excitotoxicity and oxidative stress, was uncommon, highlighting a critical research gap that requires further investigation. Discussed in detailed sections below and summarized in Table 1 are these studies and their specific investigations of DSCs in SCI and their effects on secondary injury.
Accumulating evidence highlights that the greatest therapeutic benefit following stem cell administration into a neurodegenerative (for a review, see [122]) or injured CNS does not come from the replenishment of neural or glial cell populations, but from the propagation of a supportive environment. Paracrine signaling and cell-to-cell interactions, in which DSCs have demonstrated superior capabilities compared to MSC populations, appear to be key to this success [123,124]. Limited direct comparisons between DSCs and other MSCs in the context of neuro-regeneration and the attenuation of secondary biochemical injury exist. Therefore, although less consequential to the application of regenerative medicine, in vitro studies are included in Table 2 to demonstrate biochemical mechanisms that are superior in DSCs compared to other stems cell types, or vice-versa; however, in vivo investigations are necessary to substantiate the claims of these studies in clinically relevant models. Despite this limitation, a distinction can be delineated, suggesting the stronger beneficial effects of DSCs over MSCs (Table 2). Due to a paucity of literature of relevance to SCI, the remainder of this review also draws on the literature from in vitro studies as well as other CNS neurotrauma, such as TBI and stroke, where relevant.

4.1. Angiogenesis

The ability of a therapy to revascularize ischemic CNS tissue is essential to provide blood supply and initiate repair of the damaged area. In vitro analyses demonstrate the extensive secretion of the angiogenic factor vascular endothelial growth factor (VEGF) [133,134] by DSCs at significantly higher levels than BMSCs [107,130]. DSC-secreted VEGF was found to induce the migration of endothelial cells towards DSCs and increase endothelial tubulogenesis in vitro within 24 h [135,136]. Dental stem cells, particularly those derived from the dental pulp, have also shown powerful angiogenic and vasculogenic potential in vivo. In an in vivo fertilized chick egg assay, DSCs increased blood vessel formation within 3 d [135,136], and increased vascularization and functional blood perfusion in new tissue growth after scaffold implantation in rats [100]. Of note, more prolific vessel formation was observed after DSC transplantation compared to BMSCs in a validated model of stroke [130]. DPSCs and MSCs were observed to localize around blood vessels in vitro [130] and in an in vivo model of SCI [137], respectively, appearing to act as stabilizing structural support cells for pericytes and other cell types involved in de novo angiogenesis.
Adipose tissue-derived MSCs (AMSCs), exosomes derived from BMSCs [137,138], and DSCs [121] have also been observed to improve the BSCB compromise that accompanies a SCI by supporting the maturation of neovascularization in rats, via a mechanism that is postulated to promote pericyte proliferation. Specifically, DSCs were found to differentiate into pericytes that could regulate vascular function to reduce hypoxia after SCI [121]. In other SCI rodent models, IV-engrafted BMSCs [139] as well as the intra-spinal engraftment of DSCs [115,116] reduced intraspinal hemorrhage, although further analysis is needed to elucidate the mechanistic basis of this attenuated hemorrhage. The intra-spinal engraftment of DSC hydrogels also stimulated angiogenesis, blood perfusion, and blood vessel organization within the intraspinal lesion epicenter in mouse and rat models [106,117,120]. After engraftment into a complete transection SCI rat model, angiogenesis and increased blood vessel density within sensory tract areas (measured by CD31 staining) were stimulated by DSC-loaded scaffolds, promoting sensory fiber regeneration and improving sensory function [100]. Of importance, studies comparing the effects of BMSCs with DSCs within a rat cerebral ischemic injury model showed increased blood vessel formation in the groups treated with DSCs, implying a mechanism of action of DSCs that is not common to MSCs [129]. Interestingly, a recent pre-clinical study reported that, whereas AMSCs secreted large amounts of various pro-angiogenic factors in vitro, including VEGF, these pro-angiogenic profiles where not maintained under in vivo conditions (PDGF-AA, endothelin-1, TIMP-1, and Serpin-E1) [137]. These data highlight the need for further in-depth histological analyses.

4.2. Anti-Excitotoxic Effects

In in vitro neuronal cultures, MSCs and DSCs have been demonstrated to confer significant protection against glutamate- and NMDA-mediated excitotoxicity. DSCs were able to increase the viability and survival rate of neurons cultured under excitotoxic conditions [124,140], and AMSCs and BMSCs restored mitochondrial function, ATP production, and NAD+/NADH mitochondrial respiration substrates as well as inhibited NMDA receptor subunit expression in neurons [141,142]. Although not extensively investigated in vivo, MSCs and DSCs are also able to reduce excitotoxicity in animal models of SCI. Watanabe et al. (2015) reported that BMSCs reduce the expression of several markers (e.g., PKC-y and p-CREB) implicated in the pathophysiology of glutamate-induced neuronal hyperexcitability and neuropathic pain in spinal neurons in mice [143], while Nishida et al. (2020) demonstrated that human umbilical cord MSCs (UMSC) protect neurons and restore function in a rat model of glutamate-induced cytotoxicity and spinal cord damage [144]. Likewise, the intra-spinal engraftment of SHED was demonstrated to reduce the overexpression of glutamate-induced neuronal nitric oxide synthase (nNOS), as well as the excitatory amino acid transporter 3 (EAAT3) to limit glutamate-mediated cytotoxicity after SCI in rats [110], and Ying et al. (2023) demonstrated a reduction in over-active glutaminergic synapses and increased GABAergic inhibitory synapses after DPSC engraftment in mice after SCI [117]. However, overall analyses into the anti-excitotoxic effects of DSCs are scarce and require further in-depth investigation.

4.3. Anti-Oxidative Effects

In culture with neurons, DSC-CM or DSCs have exhibited the ability to reduce DNA damage, ROS, and NO after oxidative stress [115,145,146,147], and survive hydrogen peroxide-induced oxidative stress with unperturbed growth factor expression [108]. Similarly, AMSC [148] and DSC [149,150] treatment improved neuronal stem cell viability and prevented apoptosis under cytotoxic oxidative conditions in vivo. Interestingly, the findings of Song et al. (2015) demonstrate the superior ability of DPSCs to reduce ischemia-induced astrocyte death in culture when compared to BMSCs [132]. In SCI animal models of stem cell engraftment, DSCs were able to reduce ROS production to counteract ROS-mediated neuroinflammation [104], reduced iNOS levels [103,107] involved in the overproduction of NO, and limited lipid peroxidation (4-HNE staining) while increasing the expression of the GPX4 anti-oxidant [117]. In animal models of other CNS injuries, DSC transplantation was shown to reduce the production of the oxidative stress markers 4-HNE [151] and 3-NT to a greater extent than BMSCs [124] and ROS [152]. A canine model of SCI likewise demonstrated the significant attenuation of 4-HNE and protein carbonyl associated lipid peroxidation following intravenous MSC infusion [153]. As noted for investigations into the anti-excitotoxic effect of stem cells, further characterization is required to deduce the specific antioxidative mechanisms of DSCs and elucidate whether they directly or indirectly affect cell function after SCI.

4.4. Neuroimmunomodulation

Perhaps the most extensively studied and potent capability of mesenchymal and dental stem cells in the context of SCI is their innate ability to ameliorate the harsh and refractory pro-inflammatory cascade. Attenuating pro-inflammation acutely to limit inflammatory dysfunction in the later stages of injury is a necessary feature for all SCI therapeutics. In in vivo models of SCI, BMSCs [154,155] and DSCs polarized macrophages into anti-inflammatory phenotypes by increasing the expression of Arg-1, CD206, and IL-10 in macrophages/resident microglia, with DSCs showing superior capabilities to achieve this when compared to BMSCs [107]. Furthermore, the in vivo engraftment of DSCs reduced the expression of pro-inflammatory iNOS, IL-6, CD16/32, and IL-1β [102] within the spinal cord lesion. Other studies showed that MSCs and DSCs alter macrophage polarization into anti-inflammatory phenotypes within 12 h of SCI engraftment [107], and can maintain an anti-inflammatory environment for up to 10 weeks [156]. Additionally, DSCs influence the inflammatory secretome that accompanies SCI, decreasing the production of the pro-inflammatory cytokines IFN-γ, TNF-α, IL-2, IL-17, IL-6, and IL-1β, normally expressed and maintained at high concentrations following injury, and concurrently increasing the secretion of the anti-inflammatory cytokines IL-4, IL-13, TGF-α, and IL-10 [96,102,107,157]. BMSCs, AMSCs [158,159], and DSCs [105] also modulate inflammasome complexes, including a reduction in NLRP3 expression and associated NF-κB-, IL-1β-, and IL-18-mediated inflammation in animal models of SCI. Importantly, DSCs conduct paracrine immunomodulation through the release of anti-inflammatory cytokines both in vitro [87,157] and in vivo, as shown for SCI models [107], and thus do not rely on cell–cell contact to induce the anti-inflammatory phenotypes of macrophages/microglia. Indeed, Matsubara et al. (2015) demonstrated the strong anti-inflammatory induction of cells in the SCI lesion following the engraftment of conditioned medium derived from SHED alone [107], while other authors found that intravenously injected BMSCs exert paracrine immunomodulatory and trophic effects upon the injured spinal cord [160]. This discovery was found despite no cells appearing to migrate to the spinal cord, which instead settle in the lungs, as revealed by bioluminescence imaging and spectrophotometric quantitation [7,161]. Although stem cell treatment has been discordantly demonstrated to both enrich [154,155] and deplete [162,163,164,165] macrophage/microglia populations acutely after SCI, stem cells ultimately generate anti-inflammatory and reparative phenotypes of immune cells. Of note, DSCs were also demonstrated to reduce microglial pyroptosis and reduce astrocytic glial scar formation in vivo [105,109,110,113]. The immunomodulatory influence of DSCs exerted upon lymphocytes was demonstrated in various in vitro models, including a reduction in the activation and proliferation of NK cells [166] and a reduction in the activation and migration of B cells [167,168], while the corresponding ratio of anti-inflammatory Treg cells was increased [128]. Evidence for the in vivo modulatory effects of DSCs on leukocytes following SCI indicates their ability to suppress T-cell infiltration [110], but a further mechanistic investigation is required.

4.5. Anti-Apoptotic Effects

Preventing or inhibiting the cascade of ongoing cell death initiated during the acute response to SCI is an important therapeutic goal. The engraftment of MSCs [162,169,170,171] and DSCs [87,106,110,112,119] has been shown to reduce the number of apoptotic neurons and glia following SCI. A possible mechanism by which stem cells exert these anti-apoptotic effects is through their inhibition of the expression of apoptosis initiating factors such as caspase-3, shown to be significantly reduced within the SCI lesion after DPSC [106] or SHED treatment [110]. Increased and decreased anti-apoptotic Bcl2 and pro-apoptotic Bax expression, respectively, were also detected in vitro [148] and in vivo in DSC-treated animal models of SCI [106].
Matsubara et al. (2015) reported the greater expression of anti-apoptotic and neuroprotective factors in SHED-CM than BMSCs, including nidogen-1, insulin, and NCAM-1 [107]. Neurotrophins play an active role in inducing inhibitors of caspase-3 [172]. Therefore, the extensive expression of neurotrophins (including BDNF, GDNF, NT-3, NGF, IGF1, and CNTF) by BMSCs [165], and superior expression by DSCs [87,90,124], may explain the mechanism of action by which stem cells reduce apoptosis [169]. Evidence is also accumulating on the effects of the stem cell regulation of autophagy following SCI, with animal models showing a promotional effect of BMSCs on autophagy through increased autophagy-related proteins beclin-1 and light chain3-II in neurons, resulting in attenuated apoptosis and improved recovery [158,173]. Overall, the literature supports the conclusion that both MSCs and DSCs can promote neuroprotection and the survival of neural and glial cells within the hostile secondary injury environment.

4.6. Tissue Preservation and Regeneration

Attenuating the secondary injury cascades described above culminates in the preservation of the gross lesion architecture and neuronal and glial connectivity. This includes decreasing the cystic cavity and lesion size and limiting glial scarring through CSPG inhibition as demonstrated by DSCs [107,111]. Improvement in the general organization of neuronal and glial structures within the lesion penumbra is also commonly demonstrated by both MSCs [174] and DSCs in vivo [97,104,106].
Improving the preservation of myelin after injury is a vital therapeutic target. MSCs improve the preservation of myelin sheaths within the lesion center as well as caudally, and increase the thickness of preserved myelin sheaths [154,156,169,174], while DSCs and DSC-CM similarly preserve white matter, increase synapse preservation, and limit oligodendrocyte cell loss after SCI [97,99]. Furthermore, DSCs have exhibited a strong capacity to promote neuritogenesis and filipodial migration towards damaged neurons in vitro [123,131], which has been substantiated in in vivo SCI models demonstrating an increase in axonal regeneration (increased acetylated tubulin, neurofilament, and neural regeneration marker GAP-43 staining) and axon preservation, neurite extension, and sprouting of serotonergic, corticospinal tract, and sensory fibers [87,95,105,106,107,108,109,120].
Following in vivo SCI engraftment, the literature has consistently shown that DSCs readily differentiate into GFAP, S-100 and APC/MBP-expressing astrocytes [97,102], Schwann cells [97], and oligodendrocytes [87], and importantly, induce neural progenitor cell proliferation [94,115,118]. While DSCs have also been demonstrated to differentiate into neuronal-like cells in vitro [88] and in vivo after SCI through neuronal marker staining [101,102,116], functional analysis of differentiated neurons is less frequently conducted to substantiate these findings. Nevertheless, as discussed in this review, the therapeutic efficacy of stem cells has been demonstrated to rely less on direct stem cell replacement, and more importantly on the protective and supportive influence of stem cells on resident and infiltrating cells and the tissue microenvironment.

5. Future Perspectives for Improving DSC Therapy Translation

5.1. Improving DSC Viability

Outside of the obvious barriers that inhibit stem cell activity in the lesion site after SCI discussed above, the translation of pre-clinical findings into human clinical trials is limited due to the low survival rate of engrafted stem cells. Most studies report either no surviving cells or survival of only <1–2% after 5–10 weeks [175,176], while many studies do not measure cell survival or migration in the first instance. Low cell survival has been postulated to be caused by persistent and damaging pro-inflammatory signaling [177,178], which sustains a harsh microenvironment at all injury stages, likely preventing immunomodulatory and reparative stem cell activity from occurring in time to limit stem cell death. Indeed, stem cells begin to die at exponential rates within hours of engraftment, [143], which has been demonstrated to negatively impact stem cell efficacy post-engraftment in neurotrauma [177,179,180], with increasing stem cell survival rate and tissue sparing strongly correlating in pre-clinical models [181]. A mechanistic understanding of the causes of stem cell death within SCI secondary injury microenvironments is lacking, which could be key to developing the timeframes and techniques aimed at ameliorating stem cell death and improving functional efficacy following engraftment. Importantly, many investigations into the protective functions of stem cells are conducted in vitro prior to their engraftment (e.g., neurotrophin secretion). A greater understanding of how the multicellular response to neurotrauma impacts these functions will be required to enhance translation in human trials. The recent interest in the therapeutic and immunomodulatory effects of apoptotic stem cells (reviewed in [182]) falls outside the scope of this review, but further highlights the need for an increased understanding into stem cell functions in various microenvironments.

5.2. Optimizing the Delivery of DSC Therapy

The intensive investigation of stem cell pharmacokinetics is another facet of attempts to achieve therapeutic efficacy. Optimizing the delivery route, timing, and dosage of stem cell treatments to improve stem cell survival and treatment efficacy remains an active goal. While no clinical trial has yet investigated the safety or efficacy of DSC engraftment protocols in participants with a primary diagnosis of spinal cord injury, various MSC phase I and phase II trials have been undertaken (reviewed in [183]). Three main routes for cell delivery are utilized in pre-clinical animal models of SCI: intraparenchymal administration via injection directly into the spinal cord tissue; intrathecal administration via lumbar puncture into the fluid-filled space surrounding the spinal cord; and intravenous administration. Xenotransplantation data for intravenous administration show that only approximately 1–2% of circulating human MSCs engraft into the hemisected spinal cord of the rat, yielding less tissue sparing and inducing more marked immunogenicity than lumbar puncture or intraspinal injection [184]. In comparison, intrathecal administration avoids the systemic circulation; however, only an approximate 8–9% increase in the number of engrafted cells is observed before further viability loss [185]. Intraparenchymal injection is more commonly utilized due to a greater control of cell localization, but cell survival remains low, likely due to immediate exposure to the lesion microenvironment [108,143]. This stereotaxic approach also poses risks to the remnant spinal tissue. Studies investigating the various timings of stem cell administration report better stem cell viability when engrafted at the 3–7 days post-injury. Engraftment occurring at chronic phases of injury also yields only weak therapeutic efficacy [143]. Importantly, no study to date has directly compared the effects of acute versus delayed engraftment on DSC viability.
Dosage is another key factor of translatability, with high inter-study variability observed within and between human and rodent investigations (reviewed in [186,187], respectively) with a higher median dosage routinely administered in animal studies (4.2 × 106/kg) than in clinical trials (1–3 × 106/kg). A recent meta-analysis found that an increased dose of intra-parenchymal transplanted stem cells (≥1 × 106 at) in the subacute phase of pre-clinical SCI (3–14 days post-SCI) had better therapeutic effects than other protocols [188]. Despite this, the majority of the DSC literature to date reports the delivery of sub-optimal doses (≤1 × 106) of DSCs immediately after surgically induced SCI (≤1 h post-SCI) (Table 1). These variables, coupled with the heterogeneity and scarcity of reported outcomes as presented in the current review, highlight future challenges in progressing the translational potential of DSC therapy to clinical trials.

5.3. Increasing Measurable Outcomes of DSC Therapy

The Basso–Beattie–Bresnahan (BBB) locomotor rating scale in rats [189] and the Basso Mouse Scale (BMS) in mice [190] are the accepted measures of motor improvement. A meta-analysis of MSC data obtained from models of SCI in the rat indicated an overall BBB improvement of 3.9 versus the controls across 83 extracted studies [187]. However, the non-linear nature of motor changes in the BBB scale and an unclear correlation to human function presents difficulties in interpreting relevance and clinical significance. Uncertainty about the clinical effectiveness of stem cell therapy is, in part, attributed to the disappointing lack of motor or sensory improvements and the overall achievement of the desired therapeutic effects. As highlighted in this review, accumulating evidence for the attenuation of secondary injury cascades in a biological response to the engraftment of exogenous stem cells, or stem cell-derived secreted factors, may offer an important adjunctive measure of stem cell effectiveness. However, the in-depth characterization and reporting of standardized biomarkers of secondary injury would be necessary in future investigations. In agreement with Shang et al. (2022), the quality of the pre-clinical literature describing stem cell delivery mode and the dosage and timing of stem cell administration represents a barrier for the field [188]. It is equally important to consider and promote the reporting of negative pre-clinical data to further solidify our understanding of the various treatment variables that are, and are not, favorable to treatment administration and limit study duplication as well as animal and resource wastage. Nevertheless, as discussed in this review, the therapeutic efficacy of stem cell therapy is the subject of active investigation and refinement. Overall, our understanding of the true potential of DSCs across the various degrees of pathological and treatment heterogeneity may be improved by the systematic reporting of a range of outcome measures, within a consensus guideline or framework.

6. Conclusions

Stem cell therapy is a promising strategy for the preservation or restoration of the structure and function of the brain and spinal cord. This review discusses the mechanisms of action of dental and mesenchymal stem cells within the CNS microenvironment during secondary degeneration and constructs a translational framework of stem cell therapies of relevance to spinal cord injury. Evidence for paracrine and cell-to-cell modulation of a range of vascular and biochemical events, inflammatory and CNS cells, their signaling pathways, and secretome is considered. We would propose that the multifunctional properties of stem cells, DSCs in particular, have a multifactorial level of control on infiltrating and resident cells and the inflammatory microenvironment that is independent of their multipotent differentiation potential.
We encourage investigations of DSCs and other stem cell therapies with increased reporting on stem cell viability and the effects (or lack thereof) on the spectrum of secondary injury mechanisms following SCI. This is vital to not only elucidate the mechanisms by which stem cells survive within and repair the harsh cytotoxic microenvironments of the injured spinal cord, but understand which variables may impact clinical translation, efficacy, and ultimately, therapeutic success.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells13100817/s1, Table S1: English search strategies for focused review.

Author Contributions

Conceptualization, S.J. and R.L.O.D.; Writing—Original Draft Preparation, S.J.; Writing—Review and Editing, S.J., R.L.O.D., J.M.C. and S.G.; Supervision, R.L.O.D., J.M.C. and S.G.; Funding Acquisition, S.J., R.L.O.D., J.M.C. and S.G.; Visualization, R.L.O.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported in part by the Neil Sachse Centre for Spinal Cord Research on behalf of its donors. S.J. was supported by an Australian Government Research Training Program (RTP) Stipend and a RTP Fee-Offset Scholarship through the University of Adelaide. This research was funded in part through the AOSpine Foundation, AOSpine (Project no AOSDIA2019-081). AOSpine is a clinical division of the AO Foundation—an independent medically-guided not-for-profit organization.

Acknowledgments

We acknowledge the late Neil Sachse (1951–2020), founder of the Neil Sachse Centre for Spinal Cord Research, SAHMRI, for his dedicated and unwavering support of our research. Figure 1, Figure 2 and Figure 3 were created using BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Middleton, J.W.; Arora, M.; Kifley, A.; Clark, J.; Borg, S.J.; Tran, Y.; Atresh, S.; Kaur, J.; Shetty, S.; Nunn, A.; et al. Australian arm of the International Spinal Cord Injury (Aus-InSCI) Community Survey: 2. Understanding the lived experience in people with spinal cord injury. Spinal Cord. 2022, 60, 1069–1079. [Google Scholar] [CrossRef]
  2. Kumar, R.; Lim, J.; Mekary, R.A.; Rattani, A.; Dewan, M.C.; Sharif, S.Y.; Osorio-Fonseca, E.; Park, K.B. Traumatic Spinal Injury: Global Epidemiology and Worldwide Volume. World Neurosurg. 2018, 113, e345–e363. [Google Scholar] [CrossRef]
  3. Krueger, H.; Noonan, V.K.; Trenaman, L.M.; Joshi, P.; Rivers, C.S. The economic burden of traumatic spinal cord injury in Canada. Chronic Dis. Inj. Can. 2013, 33, 113–122. [Google Scholar] [CrossRef]
  4. Alizadeh, A.; Dyck, S.M.; Karimi-Abdolrezaee, S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front. Neurol. 2019, 10, 282. [Google Scholar] [CrossRef]
  5. Ahuja, C.S.; Wilson, J.R.; Nori, S.; Kotter, M.R.N.; Druschel, C.; Curt, A.; Fehlings, M.G. Traumatic spinal cord injury. Nat. Rev. Dis. Primers 2017, 3, 17018. [Google Scholar] [CrossRef]
  6. Losey, P.; Young, C.; Krimholtz, E.; Bordet, R.; Anthony, D.C. The role of hemorrhage following spinal-cord injury. Brain Res. 2014, 1569, 9–18. [Google Scholar] [CrossRef]
  7. Matsushita, T.; Lankford, K.L.; Arroyo, E.J.; Sasaki, M.; Neyazi, M.; Radtke, C.; Kocsis, J.D. Diffuse and persistent blood-spinal cord barrier disruption after contusive spinal cord injury rapidly recovers following intravenous infusion of bone marrow mesenchymal stem cells. Exp. Neurol. 2015, 267, 152–164. [Google Scholar] [CrossRef]
  8. Maikos, J.T.; Shreiber, D.I. Immediate damage to the blood-spinal cord barrier due to mechanical trauma. J. Neurotrauma 2007, 24, 492–507. [Google Scholar] [CrossRef]
  9. Popovich, P.G.; Horner, P.J.; Mullin, B.B.; Stokes, B.T. A quantitative spatial analysis of the blood-spinal cord barrier. I. Permeability changes after experimental spinal contusion injury. Exp. Neurol. 1996, 142, 258–275. [Google Scholar] [CrossRef]
  10. Whetstone, W.D.; Hsu, J.Y.; Eisenberg, M.; Werb, Z.; Noble-Haeusslein, L.J. Blood-spinal cord barrier after spinal cord injury: Relation to revascularization and wound healing. J. Neurosci. Res. 2003, 74, 227–239. [Google Scholar] [CrossRef]
  11. Orem, B.C.; Rajaee, A.; Stirling, D.P. IP(3)R-mediated intra-axonal Ca(2+) release contributes to secondary axonal degeneration following contusive spinal cord injury. Neurobiol. Dis. 2020, 146, 105123. [Google Scholar] [CrossRef]
  12. Figley, S.A.; Khosravi, R.; Legasto, J.M.; Tseng, Y.F.; Fehlings, M.G. Characterization of vascular disruption and blood-spinal cord barrier permeability following traumatic spinal cord injury. J. Neurotrauma 2014, 31, 541–552. [Google Scholar] [CrossRef]
  13. Mautes, A.E.; Weinzierl, M.R.; Donovan, F.; Noble, L.J. Vascular events after spinal cord injury: Contribution to secondary pathogenesis. Phys. Ther. 2000, 80, 673–687. [Google Scholar] [CrossRef]
  14. Sekhon, L.H.; Fehlings, M.G. Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine 2001, 26, S2–S12. [Google Scholar] [CrossRef]
  15. Li, Y.; Lucas-Osma, A.M.; Black, S.; Bandet, M.V.; Stephens, M.J.; Vavrek, R.; Sanelli, L.; Fenrich, K.K.; Di Narzo, A.F.; Dracheva, S.; et al. Pericytes impair capillary blood flow and motor function after chronic spinal cord injury. Nat. Med. 2017, 23, 733–741. [Google Scholar] [CrossRef]
  16. Matute, C.; Alberdi, E.; Ibarretxe, G.; Sánchez-Gómez, M.V. Excitotoxicity in glial cells. Eur. J. Pharmacol. 2002, 447, 239–246. [Google Scholar] [CrossRef]
  17. Dong, H.W.; Hayar, A.; Callaway, J.; Yang, X.H.; Nai, Q.; Ennis, M. Group I mGluR activation enhances Ca(2+)-dependent nonselective cation currents and rhythmic bursting in main olfactory bulb external tufted cells. J. Neurosci. 2009, 29, 11943–11953. [Google Scholar] [CrossRef]
  18. Xu, G.Y.; Liu, S.; Hughes, M.G.; McAdoo, D.J. Glutamate-induced losses of oligodendrocytes and neurons and activation of caspase-3 in the rat spinal cord. Neuroscience 2008, 153, 1034–1047. [Google Scholar] [CrossRef]
  19. Mody, I.; MacDonald, J.F. NMDA receptor-dependent excitotoxicity: The role of intracellular Ca2+ release. Trends Pharmacol. Sci. 1995, 16, 356–359. [Google Scholar] [CrossRef]
  20. O’Hare Doig, R.L.; Santhakumar, S.; Fehily, B.; Raja, S.; Solomon, T.; Bartlett, C.A.; Fitzgerald, M.; Hodgetts, S.I. Acute Cellular and Functional Changes With a Combinatorial Treatment of Ion Channel Inhibitors Following Spinal Cord Injury. Front. Mol. Neurosci. 2020, 13, 85. [Google Scholar] [CrossRef]
  21. Schmidt, J.; Quintá, H.R. Mitochondrial dysfunction as a target in spinal cord injury: Intimate correlation between pathological processes and therapeutic approaches. Neural Regen. Res. 2023, 18, 2161–2166. [Google Scholar] [CrossRef]
  22. Vaishnav, R.A.; Singh, I.N.; Miller, D.M.; Hall, E.D. Lipid peroxidation-derived reactive aldehydes directly and differentially impair spinal cord and brain mitochondrial function. J. Neurotrauma 2010, 27, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
  23. Bastani, N.E.; Kostovski, E.; Sakhi, A.K.; Karlsen, A.; Carlsen, M.H.; Hjeltnes, N.; Blomhoff, R.; Iversen, P.O. Reduced antioxidant defense and increased oxidative stress in spinal cord injured patients. Arch. Phys. Med. Rehabil. 2012, 93, 2223–2228. [Google Scholar] [CrossRef] [PubMed]
  24. Hall, E.D.; Wang, J.A.; Bosken, J.M.; Singh, I.N. Lipid peroxidation in brain or spinal cord mitochondria after injury. J. Bioenerg. Biomembr. 2016, 48, 169–174. [Google Scholar] [CrossRef] [PubMed]
  25. Sullivan, P.G.; Krishnamurthy, S.; Patel, S.P.; Pandya, J.D.; Rabchevsky, A.G. Temporal characterization of mitochondrial bioenergetics after spinal cord injury. J. Neurotrauma 2007, 24, 991–999. [Google Scholar] [CrossRef] [PubMed]
  26. Visavadiya, N.P.; Patel, S.P.; VanRooyen, J.L.; Sullivan, P.G.; Rabchevsky, A.G. Cellular and subcellular oxidative stress parameters following severe spinal cord injury. Redox Biol. 2016, 8, 59–67. [Google Scholar] [CrossRef] [PubMed]
  27. Huntemer-Silveira, A.; Patil, N.; Brickner, M.A.; Parr, A.M. Strategies for Oligodendrocyte and Myelin Repair in Traumatic CNS Injury. Front. Cell Neurosci. 2020, 14, 619707. [Google Scholar] [CrossRef] [PubMed]
  28. Fleming, J.C.; Norenberg, M.D.; Ramsay, D.A.; Dekaban, G.A.; Marcillo, A.E.; Saenz, A.D.; Pasquale-Styles, M.; Dietrich, W.D.; Weaver, L.C. The cellular inflammatory response in human spinal cords after injury. Brain 2006, 129, 3249–3269. [Google Scholar] [CrossRef]
  29. Casella, G.T.; Bunge, M.B.; Wood, P.M. Endothelial cell loss is not a major cause of neuronal and glial cell death following contusion injury of the spinal cord. Exp. Neurol. 2006, 202, 8–20. [Google Scholar] [CrossRef]
  30. Springer, J.E.; Azbill, R.D.; Knapp, P.E. Activation of the caspase-3 apoptotic cascade in traumatic spinal cord injury. Nat. Med. 1999, 5, 943–946. [Google Scholar] [CrossRef]
  31. Chen, K.B.; Uchida, K.; Nakajima, H.; Yayama, T.; Hirai, T.; Watanabe, S.; Guerrero, A.R.; Kobayashi, S.; Ma, W.Y.; Liu, S.Y.; et al. Tumor necrosis factor-α antagonist reduces apoptosis of neurons and oligodendroglia in rat spinal cord injury. Spine 2011, 36, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
  32. Kotipatruni, R.R.; Dasari, V.R.; Veeravalli, K.K.; Dinh, D.H.; Fassett, D.; Rao, J.S. p53- and Bax-mediated apoptosis in injured rat spinal cord. Neurochem. Res. 2011, 36, 2063–2074. [Google Scholar] [CrossRef] [PubMed]
  33. Guha, L.; Singh, N.; Kumar, H. Different Ways to Die: Cell Death Pathways and Their Association With Spinal Cord Injury. Neurospine 2023, 20, 430–448. [Google Scholar] [CrossRef] [PubMed]
  34. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef] [PubMed]
  35. Kigerl, K.A.; Gensel, J.C.; Ankeny, D.P.; Alexander, J.K.; Donnelly, D.J.; Popovich, P.G. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. Neurosci. 2009, 29, 13435–13444. [Google Scholar] [CrossRef] [PubMed]
  36. Pineau, I.; Lacroix, S. Proinflammatory cytokine synthesis in the injured mouse spinal cord: Multiphasic expression pattern and identification of the cell types involved. J. Comp. Neurol. 2007, 500, 267–285. [Google Scholar] [CrossRef] [PubMed]
  37. Hellenbrand, D.J.; Quinn, C.M.; Piper, Z.J.; Elder, R.T.; Mishra, R.R.; Marti, T.L.; Omuro, P.M.; Roddick, R.M.; Lee, J.S.; Murphy, W.L.; et al. The secondary injury cascade after spinal cord injury: An analysis of local cytokine/chemokine regulation. Neural Regen. Res. 2024, 19, 1308–1317. [Google Scholar] [CrossRef] [PubMed]
  38. Rosas Almanza, J.; Stehlik, K.E.; Page, J.J.; Xiong, S.H.; Tabor, E.G.; Aperi, B.; Patel, K.; Kodali, R.; Kurpad, S.; Budde, M.D.; et al. IL-12p40 promotes secondary damage and functional impairment after spinal cord contusional injury. J. Neurosci. Res. 2022, 100, 2213–2231. [Google Scholar] [CrossRef]
  39. Schnell, L.; Schneider, R.; Berman, M.A.; Perry, V.H.; Schwab, M.E. Lymphocyte recruitment following spinal cord injury in mice is altered by prior viral exposure. Eur. J. Neurosci. 1997, 9, 1000–1007. [Google Scholar] [CrossRef]
  40. Greenhalgh, A.D.; David, S. Differences in the phagocytic response of microglia and peripheral macrophages after spinal cord injury and its effects on cell death. J. Neurosci. 2014, 34, 6316–6322. [Google Scholar] [CrossRef]
  41. Wanner, I.B.; Anderson, M.A.; Song, B.; Levine, J.; Fernandez, A.; Gray-Thompson, Z.; Ao, Y.; Sofroniew, M.V. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J. Neurosci. 2013, 33, 12870–12886. [Google Scholar] [CrossRef]
  42. Schnell, L.; Fearn, S.; Klassen, H.; Schwab, M.E.; Perry, V.H. Acute inflammatory responses to mechanical lesions in the CNS: Differences between brain and spinal cord. Eur. J. Neurosci. 1999, 11, 3648–3658. [Google Scholar] [CrossRef] [PubMed]
  43. Bresnahan, J.C. An electron-microscopic analysis of axonal alterations following blunt contusion of the spinal cord of the rhesus monkey (Macaca mulatta). J. Neurol. Sci. 1978, 37, 59–82. [Google Scholar] [CrossRef] [PubMed]
  44. Williams, P.R.; Marincu, B.N.; Sorbara, C.D.; Mahler, C.F.; Schumacher, A.M.; Griesbeck, O.; Kerschensteiner, M.; Misgeld, T. A recoverable state of axon injury persists for hours after spinal cord contusion in vivo. Nat. Commun. 2014, 5, 5683. [Google Scholar] [CrossRef] [PubMed]
  45. Fehlings, M.G.; Tator, C.H. The relationships among the severity of spinal cord injury, residual neurological function, axon counts, and counts of retrogradely labeled neurons after experimental spinal cord injury. Exp. Neurol. 1995, 132, 220–228. [Google Scholar] [CrossRef] [PubMed]
  46. Soderblom, C.; Luo, X.; Blumenthal, E.; Bray, E.; Lyapichev, K.; Ramos, J.; Krishnan, V.; Lai-Hsu, C.; Park, K.K.; Tsoulfas, P.; et al. Perivascular fibroblasts form the fibrotic scar after contusive spinal cord injury. J. Neurosci. 2013, 33, 13882–13887. [Google Scholar] [CrossRef] [PubMed]
  47. Sharma, K.; Selzer, M.E.; Li, S. Scar-mediated inhibition and CSPG receptors in the CNS. Exp. Neurol. 2012, 237, 370–378. [Google Scholar] [CrossRef]
  48. Gonzenbach, R.R.; Schwab, M.E. Disinhibition of neurite growth to repair the injured adult CNS: Focusing on Nogo. Cell Mol. Life Sci. 2008, 65, 161–176. [Google Scholar] [CrossRef]
  49. Monnier, P.P.; Sierra, A.; Schwab, J.M.; Henke-Fahle, S.; Mueller, B.K. The Rho/ROCK pathway mediates neurite growth-inhibitory activity associated with the chondroitin sulfate proteoglycans of the CNS glial scar. Mol. Cell Neurosci. 2003, 22, 319–330. [Google Scholar] [CrossRef]
  50. Duncan, G.J.; Manesh, S.B.; Hilton, B.J.; Assinck, P.; Plemel, J.R.; Tetzlaff, W. The fate and function of oligodendrocyte progenitor cells after traumatic spinal cord injury. Glia 2020, 68, 227–245. [Google Scholar] [CrossRef]
  51. Ankeny, D.P.; Lucin, K.M.; Sanders, V.M.; McGaughy, V.M.; Popovich, P.G. Spinal cord injury triggers systemic autoimmunity: Evidence for chronic B lymphocyte activation and lupus-like autoantibody synthesis. J. Neurochem. 2006, 99, 1073–1087. [Google Scholar] [CrossRef] [PubMed]
  52. Beattie, M.S.; Hermann, G.E.; Rogers, R.C.; Bresnahan, J.C. Cell death in models of spinal cord injury. Prog. Brain Res. 2002, 137, 37–47. [Google Scholar] [CrossRef] [PubMed]
  53. Rowland, J.W.; Hawryluk, G.W.; Kwon, B.; Fehlings, M.G. Current status of acute spinal cord injury pathophysiology and emerging therapies: Promise on the horizon. Neurosurg. Focus 2008, 25, E2. [Google Scholar] [CrossRef] [PubMed]
  54. Sharif-Alhoseini, M.; Khormali, M.; Rezaei, M.; Safdarian, M.; Hajighadery, A.; Khalatbari, M.M.; Safdarian, M.; Meknatkhah, S.; Rezvan, M.; Chalangari, M.; et al. Animal models of spinal cord injury: A systematic review. Spinal Cord. 2017, 55, 714–721. [Google Scholar] [CrossRef] [PubMed]
  55. Estrada, V.; Oldenburg, E.; Popa, O.; Müller, H.W. Mapping the Long Rocky Road to Effective Spinal Cord Injury Therapy: A Meta-Review of Pre-Clinical and Clinical Research. J. Neurotrauma 2022, 39, 591–612. [Google Scholar] [CrossRef] [PubMed]
  56. Varma, A.K.; Das, A.; Wallace, G.; Barry, J.; Vertegel, A.A.; Ray, S.K.; Banik, N.L. Spinal Cord Injury: A Review of Current Therapy, Future Treatments, and Basic Science Frontiers. Neurochem. Res. 2013, 38, 895–905. [Google Scholar] [CrossRef] [PubMed]
  57. Bracken, M.B. Methylprednisolone and acute spinal cord injury: An update of the randomized evidence. Spine 2001, 26, S47–S54. [Google Scholar] [CrossRef]
  58. Fehlings, M.G.; Moghaddamjou, A.; Harrop, J.S.; Stanford, R.; Ball, J.; Aarabi, B.; Freeman, B.J.C.; Arnold, P.M.; Guest, J.D.; Kurpad, S.N.; et al. Safety and Efficacy of Riluzole in Acute Spinal Cord Injury Study (RISCIS): A Multi-Center, Randomized, Placebo-Controlled, Double-Blinded Trial. J. Neurotrauma 2023, 40, 1878–1888. [Google Scholar] [CrossRef]
  59. Zhang, Y.; Al Mamun, A.; Yuan, Y.; Lu, Q.; Xiong, J.; Yang, S.; Wu, C.; Wu, Y.; Wang, J. Acute spinal cord injury: Pathophysiology and pharmacological intervention (Review). Mol. Med. Rep. 2021, 23, 417. [Google Scholar] [CrossRef]
  60. Meletis, K.; Barnabé-Heider, F.; Carlén, M.; Evergren, E.; Tomilin, N.; Shupliakov, O.; Frisén, J. Spinal cord injury reveals multilineage differentiation of ependymal cells. PLoS Biol. 2008, 6, e182. [Google Scholar] [CrossRef]
  61. Hamilton, L.K.; Truong, M.K.V.; Bednarczyk, M.R.; Aumont, A.; Fernandes, K.J.L. Cellular organization of the central canal ependymal zone, a niche of latent neural stem cells in the adult mammalian spinal cord. Neuroscience 2009, 164, 1044–1056. [Google Scholar] [CrossRef]
  62. Marichal, N.; García, G.; Radmilovich, M.; Trujillo-Cenóz, O.; Russo, R.E. Spatial Domains of Progenitor-Like Cells and Functional Complexity of a Stem Cell Niche in the Neonatal Rat Spinal Cord. Stem Cells 2012, 30, 2020–2031. [Google Scholar] [CrossRef]
  63. Torrillas de la Cal, A.; Paniagua-Torija, B.; Arevalo-Martin, A.; Faulkes, C.G.; Jiménez, A.J.; Ferrer, I.; Molina-Holgado, E.; Garcia-Ovejero, D. The Structure of the Spinal Cord Ependymal Region in Adult Humans Is a Distinctive Trait among Mammals. Cells 2021, 10, 2235. [Google Scholar] [CrossRef]
  64. Paniagua-Torija, B.; Norenberg, M.; Arevalo-Martin, A.; Carballosa-Gautam, M.M.; Campos-Martin, Y.; Molina-Holgado, E.; Garcia-Ovejero, D. Cells in the adult human spinal cord ependymal region do not proliferate after injury. J. Pathol. 2018, 246, 415–421. [Google Scholar] [CrossRef]
  65. Huang, L.; Fu, C.; Xiong, F.; He, C.; Wei, Q. Stem Cell Therapy for Spinal Cord Injury. Cell Transplant. 2021, 30, 0963689721989266. [Google Scholar] [CrossRef]
  66. Wang, Y.; Yi, H.; Song, Y. The safety of MSC therapy over the past 15 years: A meta-analysis. Stem Cell Res. Ther. 2021, 12, 545. [Google Scholar] [CrossRef]
  67. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  68. Ghaneialvar, H.; Soltani, L.; Rahmani, H.R.; Lotfi, A.S.; Soleimani, M. Characterization and Classification of Mesenchymal Stem Cells in Several Species Using Surface Markers for Cell Therapy Purposes. Indian J. Clin. Biochem. 2018, 33, 46–52. [Google Scholar] [CrossRef]
  69. Yang, Y.H.; Lee, A.J.; Barabino, G.A. Coculture-driven mesenchymal stem cell-differentiated articular chondrocyte-like cells support neocartilage development. Stem Cells Transl. Med. 2012, 1, 843–854. [Google Scholar] [CrossRef] [PubMed]
  70. Witt, R.; Weigand, A.; Boos, A.M.; Cai, A.; Dippold, D.; Boccaccini, A.R.; Schubert, D.W.; Hardt, M.; Lange, C.; Arkudas, A.; et al. Mesenchymal stem cells and myoblast differentiation under HGF and IGF-1 stimulation for 3D skeletal muscle tissue engineering. BMC Cell Biol. 2017, 18, 15. [Google Scholar] [CrossRef] [PubMed]
  71. Friedman, M.S.; Long, M.W.; Hankenson, K.D. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J. Cell Biochem. 2006, 98, 538–554. [Google Scholar] [CrossRef]
  72. Bueno, C.; Martínez-Morga, M.; García-Bernal, D.; Moraleda, J.M.; Martínez, S. Differentiation of human adult-derived stem cells towards a neural lineage involves a dedifferentiation event prior to differentiation to neural phenotypes. Scientific Reports 2021, 11, 12034. [Google Scholar] [CrossRef]
  73. Pelegri, N.G.; Milthorpe, B.K.; Gorrie, C.A.; Santos, J. Neurogenic marker expression in differentiating human adipose derived adult mesenchymal stem cells. Stem Cell Investig. 2023, 10. [Google Scholar] [CrossRef]
  74. Bertani, N.; Malatesta, P.; Volpi, G.; Sonego, P.; Perris, R. Neurogenic potential of human mesenchymal stem cells revisited: Analysis by immunostaining, time-lapse video and microarray. J. Cell Sci. 2005, 118, 3925–3936. [Google Scholar] [CrossRef]
  75. McCormick, J.B.; Huso, H.A. Stem cells and ethics: Current issues. J. Cardiovasc. Transl. Res. 2010, 3, 122–127. [Google Scholar] [CrossRef] [PubMed]
  76. 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]
  77. von Bahr, L.; Batsis, I.; Moll, G.; Hägg, M.; Szakos, A.; Sundberg, B.; Uzunel, M.; Ringden, O.; Le Blanc, K. Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells 2012, 30, 1575–1578. [Google Scholar] [CrossRef]
  78. Gu, L.-H.; Zhang, T.-T.; Li, Y.; Yan, H.-J.; Qi, H.; Li, F.-R. Immunogenicity of allogeneic mesenchymal stem cells transplanted via different routes in diabetic rats. Cell. Mol. Immunol. 2015, 12, 444–455. [Google Scholar] [CrossRef]
  79. Le Blanc, K.; Tammik, C.; Rosendahl, K.; Zetterberg, E.; Ringdén, O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp. Hematol. 2003, 31, 890–896. [Google Scholar] [CrossRef] [PubMed]
  80. Sivanathan, K.N.; Gronthos, S.; Rojas-Canales, D.; Thierry, B.; Coates, P.T. Interferon-gamma modification of mesenchymal stem cells: Implications of autologous and allogeneic mesenchymal stem cell therapy in allotransplantation. Stem Cell Rev. Rep. 2014, 10, 351–375. [Google Scholar] [CrossRef] [PubMed]
  81. Shi, S.; Robey, P.G.; Gronthos, S. Comparison of human dental pulp and bone marrow stromal stem cells by cDNA microarray analysis. Bone 2001, 29, 532–539. [Google Scholar] [CrossRef]
  82. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef]
  83. Huang, G.T.; Gronthos, S.; Shi, S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: Their biology and role in regenerative medicine. J. Dent. Res. 2009, 88, 792–806. [Google Scholar] [CrossRef]
  84. Luo, L.; He, Y.; Wang, X.; Key, B.; Lee, B.H.; Li, H.; Ye, Q. Potential Roles of Dental Pulp Stem Cells in Neural Regeneration and Repair. Stem Cells Int. 2018, 2018, 1731289. [Google Scholar] [CrossRef]
  85. Gronthos, S.; Brahim, J.; Li, W.; Fisher, L.W.; Cherman, N.; Boyde, A.; DenBesten, P.; Robey, P.G.; Shi, S. Stem cell properties of human dental pulp stem cells. J. Dent. Res. 2002, 81, 531–535. [Google Scholar] [CrossRef]
  86. Al-Maswary, A.A.; O’Reilly, M.; Holmes, A.P.; Walmsley, A.D.; Cooper, P.R.; Scheven, B.A. Exploring the neurogenic differentiation of human dental pulp stem cells. PLoS ONE 2022, 17, e0277134. [Google Scholar] [CrossRef]
  87. Sakai, K.; Yamamoto, A.; Matsubara, K.; Nakamura, S.; Naruse, M.; Yamagata, M.; Sakamoto, K.; Tauchi, R.; Wakao, N.; Imagama, S.; et al. Human dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat spinal cord by multiple neuro-regenerative mechanisms. J. Clin. Investig. 2012, 122, 80–90. [Google Scholar] [CrossRef]
  88. Kumar, A.; Kumar, V.; Rattan, V.; Jha, V.; Bhattacharyya, S. Secretome Cues Modulate the Neurogenic Potential of Bone Marrow and Dental Stem Cells. Mol. Neurobiol. 2017, 54, 4672–4682. [Google Scholar] [CrossRef]
  89. Arthur, A.; Rychkov, G.; Shi, S.; Koblar, S.A.; Gronthos, S. Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues. Stem Cells 2008, 26, 1787–1795. [Google Scholar] [CrossRef] [PubMed]
  90. Pagella, P.; Miran, S.; Neto, E.; Martin, I.; Lamghari, M.; Mitsiadis, T.A. Human dental pulp stem cells exhibit enhanced properties in comparison to human bone marrow stem cells on neurites outgrowth. FASEB J. 2020, 34, 5499–5511. [Google Scholar] [CrossRef] [PubMed]
  91. Gronthos, S.; Arthur, A.; Bartold, P.M.; Shi, S. A method to isolate and culture expand human dental pulp stem cells. Methods Mol. Biol. 2011, 698, 107–121. [Google Scholar] [CrossRef]
  92. Kang, Y.H.; Lee, H.J.; Jang, S.J.; Byun, J.H.; Lee, J.S.; Lee, H.C.; Park, W.U.; Lee, J.H.; Rho, G.J.; Park, B.W. Immunomodulatory properties and in vivo osteogenesis of human dental stem cells from fresh and cryopreserved dental follicles. Differentiation 2015, 90, 48–58. [Google Scholar] [CrossRef]
  93. Xavier Acasigua, G.A.; Bernardi, L.; Braghirolli, D.I.; Filho, M.S.; Pranke, P.; Medeiros Fossati, A.C. Nanofiber scaffolds support bone regeneration associated with pulp stem cells. Curr. Stem Cell Res. Ther. 2014, 9, 330–337. [Google Scholar] [CrossRef] [PubMed]
  94. Nicola, F.; Marques, M.R.; Odorcyk, F.; Petenuzzo, L.; Aristimunha, D.; Vizuete, A.; Sanches, E.F.; Pereira, D.P.; Maurmann, N.; Gonçalves, C.A.; et al. Stem Cells from Human Exfoliated Deciduous Teeth Modulate Early Astrocyte Response after Spinal Cord Contusion. Mol. Neurobiol. 2019, 56, 748–760. [Google Scholar] [CrossRef] [PubMed]
  95. Albashari, A.; He, Y.; Zhang, Y.; Ali, J.; Lin, F.; Zheng, Z.; Zhang, K.; Cao, Y.; Xu, C.; Luo, L.; et al. Thermosensitive bFGF-Modified Hydrogel with Dental Pulp Stem Cells on Neuroinflammation of Spinal Cord Injury. ACS Omega 2020, 5, 16064–16075. [Google Scholar] [CrossRef]
  96. Albashari, A.A.; He, Y.; Luo, Y.; Duan, X.; Ali, J.; Li, M.; Fu, D.; Xiang, Y.; Peng, Y.; Li, S.; et al. Local Spinal Cord Injury Treatment Using a Dental Pulp Stem Cell Encapsulated H(2) S Releasing Multifunctional Injectable Hydrogel. Adv. Heal. Mater. 2023, 13, e2302286. [Google Scholar] [CrossRef]
  97. de Almeida, F.M.; Marques, S.A.; Ramalho Bdos, S.; Rodrigues, R.F.; Cadilhe, D.V.; Furtado, D.; Kerkis, I.; Pereira, L.V.; Rehen, S.K.; Martinez, A.M. Human dental pulp cells: A new source of cell therapy in a mouse model of compressive spinal cord injury. J. Neurotrauma 2011, 28, 1939–1949. [Google Scholar] [CrossRef] [PubMed]
  98. Asadi-Golshan, R.; Razban, V.; Mirzaei, E.; Rahmanian, A.; Khajeh, S.; Mostafavi-Pour, Z.; Dehghani, F. Sensory and Motor Behavior Evidences Supporting the Usefulness of Conditioned Medium from Dental Pulp-Derived Stem Cells in Spinal Cord Injury in Rats. Asian Spine J. 2018, 12, 785–793. [Google Scholar] [CrossRef] [PubMed]
  99. Asadi-Golshan, R.; Razban, V.; Mirzaei, E.; Rahmanian, A.; Khajeh, S.; Mostafavi-Pour, Z.; Dehghani, F. Efficacy of dental pulp-derived stem cells conditioned medium loaded in collagen hydrogel in spinal cord injury in rats: Stereological evidence. J. Chem. Neuroanat. 2021, 116, 101978. [Google Scholar] [CrossRef]
  100. Guo, S.; Redenski, I.; Landau, S.; Szklanny, A.; Merdler, U.; Levenberg, S. Prevascularized Scaffolds Bearing Human Dental Pulp Stem Cells for Treating Complete Spinal Cord Injury. Adv. Health Mater. 2020, 9, e2000974. [Google Scholar] [CrossRef]
  101. Hu, Z.B.; Chen, H.C.; Wei, B.; Zhang, Z.M.; Wu, S.K.; Sun, J.C.; Xiang, M. Platelet rich plasma enhanced neuro-regeneration of human dental pulp stem cells in vitro and in rat spinal cord. Ann. Transl. Med. 2022, 10, 584. [Google Scholar] [CrossRef] [PubMed]
  102. Kabatas, S.; Demir, C.S.; Civelek, E.; Yilmaz, I.; Kircelli, A.; Yilmaz, C.; Akyuva, Y.; Karaoz, E. Neuronal regeneration in injured rat spinal cord after human dental pulp derived neural crest stem cell transplantation. Bratisl. Lek. Listy 2018, 119, 143–151. [Google Scholar] [CrossRef] [PubMed]
  103. Kandalam, S.; De Berdt, P.; Ucakar, B.; Vanvarenberg, K.; Bouzin, C.; Gratpain, V.; Diogenes, A.; Montero-Menei, C.N.; des Rieux, A. Human dental stem cells of the apical papilla associated to BDNF-loaded pharmacologically active microcarriers (PAMs) enhance locomotor function after spinal cord injury. Int. J. Pharm. 2020, 587, 119685. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, C.; Hu, F.; Jiao, G.; Guo, Y.; Zhou, P.; Zhang, Y.; Zhang, Z.; Yi, J.; You, Y.; Li, Z.; et al. Dental pulp stem cell-derived exosomes suppress M1 macrophage polarization through the ROS-MAPK-NFκB P65 signaling pathway after spinal cord injury. J. Nanobiotechnol. 2022, 20, 65. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, T.; Ma, Z.; Liu, L.; Pei, Y.; Wu, Q.; Xu, S.; Liu, Y.; Ding, N.; Guan, Y.; Zhang, Y.; et al. Conditioned medium from human dental pulp stem cells treats spinal cord injury by inhibiting microglial pyroptosis. Neural Regen. Res. 2024, 19, 1105–1111. [Google Scholar] [CrossRef]
  106. Luo, L.; Albashari, A.A.; Wang, X.; Jin, L.; Zhang, Y.; Zheng, L.; Xia, J.; Xu, H.; Zhao, Y.; Xiao, J.; et al. Effects of Transplanted Heparin-Poloxamer Hydrogel Combining Dental Pulp Stem Cells and bFGF on Spinal Cord Injury Repair. Stem Cells Int. 2018, 2018, 2398521. [Google Scholar] [CrossRef]
  107. Matsubara, K.; Matsushita, Y.; Sakai, K.; Kano, F.; Kondo, M.; Noda, M.; Hashimoto, N.; Imagama, S.; Ishiguro, N.; Suzumura, A.; et al. Secreted ectodomain of sialic acid-binding Ig-like lectin-9 and monocyte chemoattractant protein-1 promote recovery after rat spinal cord injury by altering macrophage polarity. J. Neurosci. 2015, 35, 2452–2464. [Google Scholar] [CrossRef]
  108. Nagashima, K.; Miwa, T.; Soumiya, H.; Ushiro, D.; Takeda-Kawaguchi, T.; Tamaoki, N.; Ishiguro, S.; Sato, Y.; Miyamoto, K.; Ohno, T.; et al. Priming with FGF2 stimulates human dental pulp cells to promote axonal regeneration and locomotor function recovery after spinal cord injury. Sci. Rep. 2017, 7, 13500. [Google Scholar] [CrossRef] [PubMed]
  109. Nicola, F.C.; Rodrigues, L.P.; Crestani, T.; Quintiliano, K.; Sanches, E.F.; Willborn, S.; Aristimunha, D.; Boisserand, L.; Pranke, P.; Netto, C.A. Human dental pulp stem cells transplantation combined with treadmill training in rats after traumatic spinal cord injury. Braz. J. Med. Biol. Res. 2016, 49, e5319. [Google Scholar] [CrossRef]
  110. Nicola, F.D.C.; Marques, M.R.; Odorcyk, F.; Arcego, D.M.; Petenuzzo, L.; Aristimunha, D.; Vizuete, A.; Sanches, E.F.; Pereira, D.P.; Maurmann, N.; et al. Neuroprotector effect of stem cells from human exfoliated deciduous teeth transplanted after traumatic spinal cord injury involves inhibition of early neuronal apoptosis. Brain Res. 2017, 1663, 95–105. [Google Scholar] [CrossRef]
  111. Nishii, T.; Osuka, K.; Nishimura, Y.; Ohmichi, Y.; Ohmichi, M.; Suzuki, C.; Nagashima, Y.; Oyama, T.; Abe, T.; Kato, H.; et al. Protective Mechanism of Stem Cells from Human Exfoliated Deciduous Teeth in Treating Spinal Cord Injury. J. Neurotrauma 2024. ahead of print. [Google Scholar] [CrossRef] [PubMed]
  112. Nosrat, I.V.; Widenfalk, J.; Olson, L.; Nosrat, C.A. Dental pulp cells produce neurotrophic factors, interact with trigeminal neurons in vitro, and rescue motoneurons after spinal cord injury. Dev. Biol. 2001, 238, 120–132. [Google Scholar] [CrossRef]
  113. Paes, S.M.; Castro, M.V.; Barbosa, R.M.; Politti Cartarozzi, L.; Coser, L.O.; Kempe, P.R.G.; Decarli, M.C.; Moraes, Â.M.; Barraviera, B.; Ferreira Júnior, R.S.; et al. Human dental pulp stem cell monolayer and spheroid therapy after spinal motor root avulsion in adult rats. Brain Res. 2023, 1802, 148229. [Google Scholar] [CrossRef] [PubMed]
  114. Taghipour, Z.; Karbalaie, K.; Kiani, A.; Niapour, A.; Bahramian, H.; Nasr-Esfahani, M.H.; Baharvand, H. Transplantation of undifferentiated and induced human exfoliated deciduous teeth-derived stem cells promote functional recovery of rat spinal cord contusion injury model. Stem Cells Dev. 2012, 21, 1794–1802. [Google Scholar] [CrossRef] [PubMed]
  115. Tao, N.; Dong, X.; Liu, C.; Lv, L.; Hu, F.; Zhang, H.; Li, X.; Geng, P.; Duan, H.; Wu, C.T.; et al. Co-overexpression of OPN, IGF-1 and CNTF augment the therapeutic effect of DPSC on spinal cord injury. Regen. Ther. 2023, 24, 651–661. [Google Scholar] [CrossRef] [PubMed]
  116. Yang, C.; Li, X.; Sun, L.; Guo, W.; Tian, W. Potential of human dental stem cells in repairing the complete transection of rat spinal cord. J. Neural Eng. 2017, 14, 026005. [Google Scholar] [CrossRef] [PubMed]
  117. Ying, Y.; Huang, Z.; Tu, Y.; Wu, Q.; Li, Z.; Zhang, Y.; Yu, H.; Zeng, A.; Huang, H.; Ye, J.; et al. A shear-thinning, ROS-scavenging hydrogel combined with dental pulp stem cells promotes spinal cord repair by inhibiting ferroptosis. Bioact. Mater. 2023, 22, 274–290. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, C.; Ye, W.; Zhao, M.; Long, L.; Xia, D.; Fan, Z. MLL1 inhibits the neurogenic potential of SCAPs by interacting with WDR5 and repressing HES1. Int. J. Oral Sci. 2023, 15, 48. [Google Scholar] [CrossRef]
  119. Zhang, J.; Lu, X.; Feng, G.; Gu, Z.; Sun, Y.; Bao, G.; Xu, G.; Lu, Y.; Chen, J.; Xu, L.; et al. Chitosan scaffolds induce human dental pulp stem cells to neural differentiation: Potential roles for spinal cord injury therapy. Cell Tissue Res. 2016, 366, 129–142. [Google Scholar] [CrossRef]
  120. Zhou, H.; Jing, S.; Xiong, W.; Zhu, Y.; Duan, X.; Li, R.; Peng, Y.; Kumeria, T.; He, Y.; Ye, Q. Metal-organic framework materials promote neural differentiation of dental pulp stem cells in spinal cord injury. J. Nanobiotechnol. 2023, 21, 316. [Google Scholar] [CrossRef]
  121. Zhu, S.; Ying, Y.; He, Y.; Zhong, X.; Ye, J.; Huang, Z.; Chen, M.; Wu, Q.; Zhang, Y.; Xiang, Z.; et al. Hypoxia response element-directed expression of bFGF in dental pulp stem cells improve the hypoxic environment by targeting pericytes in SCI rats. Bioact. Mater. 2021, 6, 2452–2466. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. 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]
  124. Mita, T.; Furukawa-Hibi, Y.; Takeuchi, H.; Hattori, H.; Yamada, K.; Hibi, H.; Ueda, M.; Yamamoto, A. Conditioned medium from the stem cells of human dental pulp improves cognitive function in a mouse model of Alzheimer’s disease. Behav. Brain Res. 2015, 293, 189–197. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, Z.; Ma, L.; Du, C.; Wang, J.; Zhang, C.; Hu, L.; Wang, S. Dental pulp stem cells accelerate wound healing through CCL2-induced M2 macrophages polarization. iScience 2023, 26, 108043. [Google Scholar] [CrossRef]
  126. Li, C.; Liu, Y.; Deng, M.; Li, J.; Li, S.; Li, X.; Zuo, Y.; Shen, C.; Wang, Y. Comparison of the therapeutic effects of mesenchymal stem cells derived from human dental pulp (DP), adipose tissue (AD), placental amniotic membrane (PM), and umbilical cord (UC) on postmenopausal osteoporosis. Front. Pharmacol. 2024, 15, 1349199. [Google Scholar] [CrossRef] [PubMed]
  127. Dai, Y.Y.; Ni, S.Y.; Ma, K.; Ma, Y.S.; Wang, Z.S.; Zhao, X.L. Stem cells from human exfoliated deciduous teeth correct the immune imbalance of allergic rhinitis via Treg cells in vivo and in vitro. Stem Cell Res. Ther. 2019, 10, 39. [Google Scholar] [CrossRef] [PubMed]
  128. Yamaza, T.; Kentaro, A.; Chen, C.; Liu, Y.; Shi, Y.; Gronthos, S.; Wang, S.; Shi, S. Immunomodulatory properties of stem cells from human exfoliated deciduous teeth. Stem Cell Res. Ther. 2010, 1, 5. [Google Scholar] [CrossRef]
  129. Song, M.; Lee, J.H.; Bae, J.; Bu, Y.; Kim, E.C. Human Dental Pulp Stem Cells Are More Effective Than Human Bone Marrow-Derived Mesenchymal Stem Cells in Cerebral Ischemic Injury. Cell Transpl. 2017, 26, 1001–1016. [Google Scholar] [CrossRef]
  130. Janebodin, K.; Zeng, Y.; Buranaphatthana, W.; Ieronimakis, N.; Reyes, M. VEGFR2-dependent angiogenic capacity of pericyte-like dental pulp stem cells. J. Dent. Res. 2013, 92, 524–531. [Google Scholar] [CrossRef]
  131. Senthilkumar, S.; Venugopal, C.; Parveen, S.; Shobha, K.; Rai, K.S.; Kutty, B.M.; Dhanushkodi, A. Remarkable migration propensity of dental pulp stem cells towards neurodegenerative milieu: An in vitro analysis. Neurotoxicology 2020, 81, 89–100. [Google Scholar] [CrossRef] [PubMed]
  132. Song, M.; Jue, S.S.; Cho, Y.A.; Kim, E.C. Comparison of the effects of human dental pulp stem cells and human bone marrow-derived mesenchymal stem cells on ischemic human astrocytes in vitro. J. Neurosci. Res. 2015, 93, 973–983. [Google Scholar] [CrossRef]
  133. Huang, A.H.; Snyder, B.R.; Cheng, P.H.; Chan, A.W. Putative dental pulp-derived stem/stromal cells promote proliferation and differentiation of endogenous neural cells in the hippocampus of mice. Stem Cells 2008, 26, 2654–2663. [Google Scholar] [CrossRef] [PubMed]
  134. Sugiyama, M.; Iohara, K.; Wakita, H.; Hattori, H.; Ueda, M.; Matsushita, K.; Nakashima, M. Dental pulp-derived CD31/CD146 side population stem/progenitor cells enhance recovery of focal cerebral ischemia in rats. Tissue Eng. Part A 2011, 17, 1303–1311. [Google Scholar] [CrossRef] [PubMed]
  135. Bronckaers, A.; Hilkens, P.; Fanton, Y.; Struys, T.; Gervois, P.; Politis, C.; Martens, W.; Lambrichts, I. Angiogenic properties of human dental pulp stem cells. PLoS ONE 2013, 8, e71104. [Google Scholar] [CrossRef]
  136. Hilkens, P.; Fanton, Y.; Martens, W.; Gervois, P.; Struys, T.; Politis, C.; Lambrichts, I.; Bronckaers, A. Pro-angiogenic impact of dental stem cells in vitro and in vivo. Stem Cell Res. 2014, 12, 778–790. [Google Scholar] [CrossRef] [PubMed]
  137. Menezes, K.; Rosa, B.G.; Freitas, C.; da Cruz, A.S.; de Siqueira Santos, R.; Nascimento, M.A.; Alves, D.V.L.; Bonamino, M.; Rossi, M.I.; Borojevic, R.; et al. Human mesenchymal stromal/stem cells recruit resident pericytes and induce blood vessels maturation to repair experimental spinal cord injury in rats. Sci. Rep. 2020, 10, 19604. [Google Scholar] [CrossRef] [PubMed]
  138. Zhou, Y.; Wen, L.L.; Li, Y.F.; Wu, K.M.; Duan, R.R.; Yao, Y.B.; Jing, L.J.; Gong, Z.; Teng, J.F.; Jia, Y.J. Exosomes derived from bone marrow mesenchymal stem cells protect the injured spinal cord by inhibiting pericyte pyroptosis. Neural Regen. Res. 2022, 17, 194–202. [Google Scholar] [CrossRef] [PubMed]
  139. Vawda, R.; Badner, A.; Hong, J.; Mikhail, M.; Lakhani, A.; Dragas, R.; Xhima, K.; Barretto, T.; Librach, C.L.; Fehlings, M.G. Early Intravenous Infusion of Mesenchymal Stromal Cells Exerts a Tissue Source Age-Dependent Beneficial Effect on Neurovascular Integrity and Neurobehavioral Recovery After Traumatic Cervical Spinal Cord Injury. Stem Cells Transl. Med. 2019, 8, 639–649. [Google Scholar] [CrossRef]
  140. Apel, C.; Forlenza, O.V.; de Paula, V.J.; Talib, L.L.; Denecke, B.; Eduardo, C.P.; Gattaz, W.F. The neuroprotective effect of dental pulp cells in models of Alzheimer’s and Parkinson’s disease. J. Neural Transm. 2009, 116, 71–78. [Google Scholar] [CrossRef]
  141. Hao, P.; Liang, Z.; Piao, H.; Ji, X.; Wang, Y.; Liu, Y.; Liu, R.; Liu, J. Conditioned medium of human adipose-derived mesenchymal stem cells mediates protection in neurons following glutamate excitotoxicity by regulating energy metabolism and GAP-43 expression. Metab. Brain Dis. 2014, 29, 193–205. [Google Scholar] [CrossRef]
  142. Voulgari-Kokota, A.; Fairless, R.; Karamita, M.; Kyrargyri, V.; Tseveleki, V.; Evangelidou, M.; Delorme, B.; Charbord, P.; Diem, R.; Probert, L. Mesenchymal stem cells protect CNS neurons against glutamate excitotoxicity by inhibiting glutamate receptor expression and function. Exp. Neurol. 2012, 236, 161–170. [Google Scholar] [CrossRef]
  143. Watanabe, S.; Uchida, K.; Nakajima, H.; Matsuo, H.; Sugita, D.; Yoshida, A.; Honjoh, K.; Johnson, W.E.; Baba, H. Early transplantation of mesenchymal stem cells after spinal cord injury relieves pain hypersensitivity through suppression of pain-related signaling cascades and reduced inflammatory cell recruitment. Stem Cells 2015, 33, 1902–1914. [Google Scholar] [CrossRef]
  144. Nishida, F.; Zappa Villar, M.F.; Zanuzzi, C.N.; Sisti, M.S.; Camiña, A.E.; Reggiani, P.C.; Portiansky, E.L. Intracerebroventricular Delivery of Human Umbilical Cord Mesenchymal Stem Cells as a Promising Therapy for Repairing the Spinal Cord Injury Induced by Kainic Acid. Stem Cell Rev. Rep. 2020, 16, 167–180. [Google Scholar] [CrossRef]
  145. Gnanasegaran, N.; Govindasamy, V.; Mani, V.; Abu Kasim, N.H. Neuroimmunomodulatory properties of DPSCs in an in vitro model of Parkinson’s disease. IUBMB Life 2017, 69, 689–699. [Google Scholar] [CrossRef]
  146. Gnanasegaran, N.; Govindasamy, V.; Simon, C.; Gan, Q.F.; Vincent-Chong, V.K.; Mani, V.; Krishnan Selvarajan, K.; Subramaniam, V.; Musa, S.; Abu Kasim, N.H. Effect of dental pulp stem cells in MPTP-induced old-aged mice model. Eur. J. Clin. Investig. 2017, 47, 403–414. [Google Scholar] [CrossRef]
  147. 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]
  148. Oh, J.S.; Kim, K.N.; An, S.S.; Pennant, W.A.; Kim, H.J.; Gwak, S.J.; Yoon, D.H.; Lim, M.H.; Choi, B.H.; Ha, Y. Cotransplantation of mouse neural stem cells (mNSCs) with adipose tissue-derived mesenchymal stem cells improves mNSC survival in a rat spinal cord injury model. Cell Transpl. 2011, 20, 837–849. [Google Scholar] [CrossRef]
  149. Fujii, H.; Matsubara, K.; Sakai, K.; Ito, M.; Ohno, K.; Ueda, M.; Yamamoto, A. Dopaminergic differentiation of stem cells from human deciduous teeth and their therapeutic benefits for Parkinsonian rats. Brain Res. 2015, 1613, 59–72. [Google Scholar] [CrossRef] [PubMed]
  150. Xiao, L.; Saiki, C.; Okamura, H. Oxidative Stress-Tolerant Stem Cells from Human Exfoliated Deciduous Teeth Decrease Hydrogen Peroxide-Induced Damage in Organotypic Brain Slice Cultures from Adult Mice. Int. J. Mol. Sci. 2019, 20, 1858. [Google Scholar] [CrossRef] [PubMed]
  151. Kitase, Y.; Sato, Y.; Ueda, K.; Suzuki, T.; Mikrogeorgiou, A.; Sugiyama, Y.; Matsubara, K.; Tsukagoshi Okabe, Y.; Shimizu, S.; Hirata, H.; et al. A Novel Treatment with Stem Cells from Human Exfoliated Deciduous Teeth for Hypoxic-Ischemic Encephalopathy in Neonatal Rats. Stem Cells Dev. 2020, 29, 63–74. [Google Scholar] [CrossRef] [PubMed]
  152. Ullah, I.; Choe, Y.H.; Khan, M.; Bharti, D.; Shivakumar, S.B.; Lee, H.J.; Son, Y.B.; Shin, Y.; Lee, S.L.; Park, B.W.; et al. Dental pulp-derived stem cells can counterbalance peripheral nerve injury-induced oxidative stress and supraspinal neuro-inflammation in rat brain. Sci. Rep. 2018, 8, 15795. [Google Scholar] [CrossRef] [PubMed]
  153. Kim, Y.; Jo, S.H.; Kim, W.H.; Kweon, O.K. Antioxidant and anti-inflammatory effects of intravenously injected adipose derived mesenchymal stem cells in dogs with acute spinal cord injury. Stem Cell Res. Ther. 2015, 6, 229. [Google Scholar] [CrossRef] [PubMed]
  154. Allahdadi, K.J.; de Santana, T.A.; Santos, G.C.; Azevedo, C.M.; Mota, R.A.; Nonaka, C.K.; Silva, D.N.; Valim, C.X.R.; Figueira, C.P.; Dos Santos, W.L.C.; et al. IGF-1 overexpression improves mesenchymal stem cell survival and promotes neurological recovery after spinal cord injury. Stem Cell Res. Ther. 2019, 10, 146. [Google Scholar] [CrossRef] [PubMed]
  155. Nakajima, H.; Uchida, K.; Guerrero, A.R.; Watanabe, S.; Sugita, D.; Takeura, N.; Yoshida, A.; Long, G.; Wright, K.T.; Johnson, W.E.; et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J. Neurotrauma 2012, 29, 1614–1625. [Google Scholar] [CrossRef]
  156. Bao, C.S.; Li, X.L.; Liu, L.; Wang, B.; Yang, F.B.; Chen, L.G. Transplantation of Human umbilical cord mesenchymal stem cells promotes functional recovery after spinal cord injury by blocking the expression of IL-7. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 6436–6447. [Google Scholar] [CrossRef]
  157. Ding, G.; Niu, J.; Liu, Y. Dental pulp stem cells suppress the proliferation of lymphocytes via transforming growth factor-β1. Hum. Cell 2015, 28, 81–90. [Google Scholar] [CrossRef] [PubMed]
  158. Gholaminejhad, M.; Jameie, S.B.; Abdi, M.; Abolhassani, F.; Mohammed, I.; Hassanzadeh, G. All-Trans Retinoic Acid-Preconditioned Mesenchymal Stem Cells Improve Motor Function and Alleviate Tissue Damage After Spinal Cord Injury by Inhibition of HMGB1/NF-κB/NLRP3 Pathway Through Autophagy Activation. J. Mol. Neurosci. 2022, 72, 947–962. [Google Scholar] [CrossRef] [PubMed]
  159. Huang, J.H.; Fu, C.H.; Xu, Y.; Yin, X.M.; Cao, Y.; Lin, F.Y. Extracellular Vesicles Derived from Epidural Fat-Mesenchymal Stem Cells Attenuate NLRP3 Inflammasome Activation and Improve Functional Recovery After Spinal Cord Injury. Neurochem. Res. 2020, 45, 760–771. [Google Scholar] [CrossRef]
  160. Kim, J.W.; Ha, K.Y.; Molon, J.N.; Kim, Y.H. Bone marrow-derived mesenchymal stem cell transplantation for chronic spinal cord injury in rats: Comparative study between intralesional and intravenous transplantation. Spine 2013, 38, E1065–E1074. [Google Scholar] [CrossRef]
  161. White, S.V.; Czisch, C.E.; Han, M.H.; Plant, C.D.; Harvey, A.R.; Plant, G.W. Intravenous Transplantation of Mesenchymal Progenitors Distribute Solely to the Lungs and Improve Outcomes in Cervical Spinal Cord Injury. Stem Cells 2016, 34, 1812–1825. [Google Scholar] [CrossRef] [PubMed]
  162. Novikova, L.N.; Brohlin, M.; Kingham, P.J.; Novikov, L.N.; Wiberg, M. Neuroprotective and growth-promoting effects of bone marrow stromal cells after cervical spinal cord injury in adult rats. Cytotherapy 2011, 13, 873–887. [Google Scholar] [CrossRef] [PubMed]
  163. Zeng, X.; Zeng, Y.S.; Ma, Y.H.; Lu, L.Y.; Du, B.L.; Zhang, W.; Li, Y.; Chan, W.Y. Bone marrow mesenchymal stem cells in a three-dimensional gelatin sponge scaffold attenuate inflammation, promote angiogenesis, and reduce cavity formation in experimental spinal cord injury. Cell Transpl. 2011, 20, 1881–1899. [Google Scholar] [CrossRef] [PubMed]
  164. Zhou, H.L.; Zhang, X.J.; Zhang, M.Y.; Yan, Z.J.; Xu, Z.M.; Xu, R.X. Transplantation of Human Amniotic Mesenchymal Stem Cells Promotes Functional Recovery in a Rat Model of Traumatic Spinal Cord Injury. Neurochem. Res. 2016, 41, 2708–2718. [Google Scholar] [CrossRef] [PubMed]
  165. Zhou, Z.; Chen, Y.; Zhang, H.; Min, S.; Yu, B.; He, B.; Jin, A. Comparison of mesenchymal stromal cells from human bone marrow and adipose tissue for the treatment of spinal cord injury. Cytotherapy 2013, 15, 434–448. [Google Scholar] [CrossRef] [PubMed]
  166. Yan, F.; Liu, O.; Zhang, H.; Zhou, Y.; Zhou, D.; Zhou, Z.; He, Y.; Tang, Z.; Wang, S. Human dental pulp stem cells regulate allogeneic NK cells’ function via induction of anti-inflammatory purinergic signalling in activated NK cells. Cell Prolif. 2019, 52, e12595. [Google Scholar] [CrossRef] [PubMed]
  167. Kwack, K.H.; Lee, J.M.; Park, S.H.; Lee, H.W. Human Dental Pulp Stem Cells Suppress Alloantigen-induced Immunity by Stimulating T Cells to Release Transforming Growth Factor Beta. J. Endod. 2017, 43, 100–108. [Google Scholar] [CrossRef]
  168. Liu, O.; Xu, J.; Ding, G.; Liu, D.; Fan, Z.; Zhang, C.; Chen, W.; Ding, Y.; Tang, Z.; Wang, S. Periodontal ligament stem cells regulate B lymphocyte function via programmed cell death protein 1. Stem Cells 2013, 31, 1371–1382. [Google Scholar] [CrossRef] [PubMed]
  169. Liu, J.; Chen, Q.; Zhang, Z.; Zheng, Y.; Sun, X.; Cao, X.; Gong, A.; Cui, Y.; He, Q.; Jiang, P. Fibrin scaffolds containing ectomesenchymal stem cells enhance behavioral and histological improvement in a rat model of spinal cord injury. Cells Tissues Organs 2013, 198, 35–46. [Google Scholar] [CrossRef]
  170. Qiu, X.C.; Jin, H.; Zhang, R.Y.; Ding, Y.; Zeng, X.; Lai, B.Q.; Ling, E.A.; Wu, J.L.; Zeng, Y.S. Donor mesenchymal stem cell-derived neural-like cells transdifferentiate into myelin-forming cells and promote axon regeneration in rat spinal cord transection. Stem Cell Res. Ther. 2015, 6, 105. [Google Scholar] [CrossRef]
  171. Spejo, A.B.; Chiarotto, G.B.; Ferreira, A.D.F.; Gomes, D.A.; Ferreira, R.S., Jr.; Barraviera, B.; Oliveira, A.L.R. Neuroprotection and immunomodulation following intraspinal axotomy of motoneurons by treatment with adult mesenchymal stem cells. J. Neuroinflamm. 2018, 15, 230. [Google Scholar] [CrossRef] [PubMed]
  172. Gu, Y.L.; Yin, L.W.; Zhang, Z.; Liu, J.; Liu, S.J.; Zhang, L.F.; Wang, T.H. Neurotrophin expression in neural stem cells grafted acutely to transected spinal cord of adult rats linked to functional improvement. Cell Mol. Neurobiol. 2012, 32, 1089–1097. [Google Scholar] [CrossRef] [PubMed]
  173. Gu, J.; Jin, Z.S.; Wang, C.M.; Yan, X.F.; Mao, Y.Q.; Chen, S. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Improves Spinal Cord Function After Injury in Rats by Activating Autophagy. Drug Des. Devel. Ther. 2020, 14, 1621–1631. [Google Scholar] [CrossRef] [PubMed]
  174. Fu, Q.; Liu, Y.; Liu, X.; Zhang, Q.; Chen, L.; Peng, J.; Ao, J.; Li, Y.; Wang, S.; Song, G.; et al. Engrafted peripheral blood-derived mesenchymal stem cells promote locomotive recovery in adult rats after spinal cord injury. Am. J. Transl. Res. 2017, 9, 3950–3966. [Google Scholar] [PubMed]
  175. Wang, J.; Wang, X.; Sun, Z.; Wang, X.; Yang, H.; Shi, S.; Wang, S. Stem cells from human-exfoliated deciduous teeth can differentiate into dopaminergic neuron-like cells. Stem Cells Dev. 2010, 19, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  176. Leong, W.K.; Henshall, T.L.; Arthur, A.; Kremer, K.L.; Lewis, M.D.; Helps, S.C.; Field, J.; Hamilton-Bruce, M.A.; Warming, S.; Manavis, J.; et al. Human adult dental pulp stem cells enhance poststroke functional recovery through non-neural replacement mechanisms. Stem Cells Transl. Med. 2012, 1, 177–187. [Google Scholar] [CrossRef] [PubMed]
  177. Tan, Y.; Uchida, K.; Nakajima, H.; Guerrero, A.R.; Watanabe, S.; Hirai, T.; Takeura, N.; Liu, S.Y.; Johnson, W.E.; Baba, H. Blockade of interleukin 6 signaling improves the survival rate of transplanted bone marrow stromal cells and increases locomotor function in mice with spinal cord injury. J. Neuropathol. Exp. Neurol. 2013, 72, 980–993. [Google Scholar] [CrossRef] [PubMed]
  178. Coyne, T.M.; Marcus, A.J.; Woodbury, D.; Black, I.B. Marrow stromal cells transplanted to the adult brain are rejected by an inflammatory response and transfer donor labels to host neurons and glia. Stem Cells 2006, 24, 2483–2492. [Google Scholar] [CrossRef] [PubMed]
  179. Ide, C.; Nakai, Y.; Nakano, N.; Seo, T.B.; Yamada, Y.; Endo, K.; Noda, T.; Saito, F.; Suzuki, Y.; Fukushima, M.; et al. Bone marrow stromal cell transplantation for treatment of sub-acute spinal cord injury in the rat. Brain Res. 2010, 1332, 32–47. [Google Scholar] [CrossRef]
  180. Wu, W.; Zhao, H.; Xie, B.; Liu, H.; Chen, Y.; Jiao, G.; Wang, H. Implanted spike wave electric stimulation promotes survival of the bone marrow mesenchymal stem cells and functional recovery in the spinal cord injured rats. Neurosci. Lett. 2011, 491, 73–78. [Google Scholar] [CrossRef]
  181. Nandoe Tewarie, R.D.; Hurtado, A.; Ritfeld, G.J.; Rahiem, S.T.; Wendell, D.F.; Barroso, M.M.; Grotenhuis, J.A.; Oudega, M. Bone marrow stromal cells elicit tissue sparing after acute but not delayed transplantation into the contused adult rat thoracic spinal cord. J. Neurotrauma 2009, 26, 2313–2322. [Google Scholar] [CrossRef] [PubMed]
  182. Kholodenko, I.V.; Kholodenko, R.V.; Majouga, A.G.; Yarygin, K.N. Apoptotic MSCs and MSC-Derived Apoptotic Bodies as New Therapeutic Tools. Curr. Issues Mol. Biol. 2022, 44, 351. [Google Scholar] [CrossRef] [PubMed]
  183. Shang, Z.; Wang, M.; Zhang, B.; Wang, X.; Wanyan, P. Clinical translation of stem cell therapy for spinal cord injury still premature: Results from a single-arm meta-analysis based on 62 clinical trials. BMC Med. 2022, 20, 284. [Google Scholar] [CrossRef]
  184. Paul, C.; Samdani, A.F.; Betz, R.R.; Fischer, I.; Neuhuber, B. Grafting of human bone marrow stromal cells into spinal cord injury: A comparison of delivery methods. Spine 2009, 34, 328–334. [Google Scholar] [CrossRef] [PubMed]
  185. Bakshi, A.; Barshinger, A.L.; Swanger, S.A.; Madhavani, V.; Shumsky, J.S.; Neuhuber, B.; Fischer, I. Lumbar puncture delivery of bone marrow stromal cells in spinal cord contusion: A novel method for minimally invasive cell transplantation. J. Neurotrauma 2006, 23, 55–65. [Google Scholar] [CrossRef] [PubMed]
  186. Muthu, S.; Jeyaraman, M.; Gulati, A.; Arora, A. Current evidence on mesenchymal stem cell therapy for traumatic spinal cord injury: Systematic review and meta-analysis. Cytotherapy 2021, 23, 186–197. [Google Scholar] [CrossRef]
  187. Oliveri, R.S.; Bello, S.; Biering-Sørensen, F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: Systematic review with meta-analyses of rat models. Neurobiol. Dis. 2014, 62, 338–353. [Google Scholar] [CrossRef] [PubMed]
  188. Shang, Z.; Wang, R.; Li, D.; Chen, J.; Zhang, B.; Wang, M.; Wang, X.; Wanyan, P. Spinal Cord Injury: A Systematic Review and Network Meta-Analysis of Therapeutic Strategies Based on 15 Types of Stem Cells in Animal Models. Front. Pharmacol. 2022, 13, 819861. [Google Scholar] [CrossRef] [PubMed]
  189. Basso, D.M.; Beattie, M.S.; Bresnahan, J.C. A sensitive and reliable locomotor rating scale for open field testing in rats. J. Neurotrauma 1995, 12, 1–21. [Google Scholar] [CrossRef]
  190. Basso, D.M.; Fisher, L.C.; Anderson, A.J.; Jakeman, L.B.; McTigue, D.M.; Popovich, P.G. Basso Mouse Scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J. Neurotrauma 2006, 23, 635–659. [Google Scholar] [CrossRef]
Figure 1. The secondary injury cascade following spinal cord injury involves acute to chronic sequalae that exacerbate the initial mechanical damage to the spinal cord.
Figure 1. The secondary injury cascade following spinal cord injury involves acute to chronic sequalae that exacerbate the initial mechanical damage to the spinal cord.
Cells 13 00817 g001
Figure 2. Dental stem cells are a group of mesenchymal-like neural crest derived cells that exhibit favorable qualities for neuro-regenerative therapies.
Figure 2. Dental stem cells are a group of mesenchymal-like neural crest derived cells that exhibit favorable qualities for neuro-regenerative therapies.
Cells 13 00817 g002
Figure 3. Dental stem cells and mesenchymal stem cells modulate the secondary injury microenvironment after spinal cord injury, attenuating numerous secondary cellular and biochemical cascades to improve functional outcomes after injury.
Figure 3. Dental stem cells and mesenchymal stem cells modulate the secondary injury microenvironment after spinal cord injury, attenuating numerous secondary cellular and biochemical cascades to improve functional outcomes after injury.
Cells 13 00817 g003
Table 1. The literature investigating DSC use in pre-clinical in vivo SCI models. The secondary injury target of each investigation is highlighted, along with specific and significant DSC activity and neurological benefit(s). ✓V indicates investigations also conducted in vitro.
Table 1. The literature investigating DSC use in pre-clinical in vivo SCI models. The secondary injury target of each investigation is highlighted, along with specific and significant DSC activity and neurological benefit(s). ✓V indicates investigations also conducted in vitro.
ReferenceDSC Type and GroupsDSC DoseDelivery MethodInjury ModelVascular EventsApoptosis and Cell LossBiochemical EventsInflammatory EventsStructural EventsSignificant DSC ActivityFunctional Benefit(s)
[95]HeP-hDPSCs, HeP-bFGF-hDPSCs1 × 106 Intra-spinal—immediateMice: Thoracic compression HeP + bFGF + DPSCs reduced pro-inflammatory factors (decreased in IL-6 and TNF-α); modulation of NF-κB; neuroprotection; promoted neurite and improved cell sprouting; increases in MAP-2 and Ace-Tubulin; nerve repairNot mentioned
[96]PF-OMSF/hDPSCs, PF-OMSF@JK2/hDPSCs1 × 106Intra-spinal—immediateRats: Thoracic compression Reduced pro-inflammatory factors (decreased in IL-6 and TNF-α); modulation of NF-κB; neuroprotection; promoted neurite and improved cell sprouting; increases in MAP-2 and Ace-TubulinNot mentioned
[97]hDPSCs8 × 105Intra-spinal—7d or 28d post-SCIMice: Thoracic compression Increased white matter sparing; increased neurotrophic factor expression, more so in 7 dpi engraftment group; improved tissue preservationImproved motor function (BMS) in both groups compared to the media control
[98]SHED-CM, SHED-CM + Col3 μL SHED-CMIntra-spinal—immediateRats: Thoracic compression Not mentionedSHED-CM alone did not have any effects; SHED-CM delivered with a collagen scaffold demonstrated locomotor, motor, sensory, and sensory–motor improvements
[99]SHED-CM, SHED-CM + Col3 μL SHED-CMIntra-spinal—immediateRats: Thoracic compression No effect of SHED-CM alone; SHED-CM delivered with collagen scaffold preserved gray and white matter, reduced lesion volume, limited neuronal cell loss, and limited oligodendrocyte cell lossNot mentioned
[100]hDPSCs + scaffold, hDPSCs + scaffold + HAMECs (prevascularized)0.45 × 106Intra-spinal—immediate Rats: Thoracic transection V
V
In vitro angiogenesis and neurogenesis; prevascularized DPSC scaffolds promote axon preservation (B3-tub), myelin deposition (MBP expression), and vessel formation and structure (CD31 expression, vessel volume, and vessel density); partially restored spinal cord microstructurePrevascularized DPSC scaffolds only improved sensory recovery and small improvement in motor recovery
[101]hDPSCs + PRP, hDPSCs2.5 × 105Intra-spinal—3d post-SCIRats: Thoracic contusion DPSCs reduced syrinx formation, apoptosis (TUNEL Assay); DPSCs survival up to 4 weeks and differentiation into neurons (GFAP and NeuN staining)Greatest motor function (BBB) improvement in hDPSC + PRP group (no comparisons)
[102]hDPSCs3 × 105Intra-spinal—immediateRats: Thoracic contusion Neural and glial cell differentiation (co-expression of GFAP, NF, nestin, BNDF, and vimentin); reduced pro-inflammatory factor expression (IL-1β, MPO, MIP2, and IL-6) and increased anti-inflammatory expression (IL-1ra and EP3)Improved motor function (BBB) in DPSC-treated group
[103]hSCAP, hSCAP + PAMs, hSCAP + BDNF-PAMs2 × 105Intra-spinal—immediateRats: Thoracic contusion Reduced CD68+ inflammation; reduced iNOS staining; GAP-43 and βIII-Tubulin axon growth; serotenergic fiber growthGreatest motor function (BBB) improvement in SCAP + BDNF-PAMs compared to both SCAP and the vehicle control
[104]hDPSC-derived exosomesN/ATail vein—30 min post-SCIMice: Thoracic contusion V
In vitro and in vivo LPS-induced ROS reduction, reduced M1 macrophage polarization and reduced P-ERK/ERK levels; in vivo reduction in M1 macrophage number; slight neuronal preservation (NF200 and NeuN staining) and histological reductions in structural damageImproved motor function (BMS) in the exosome-treated group
[105]hDPSCs200μL DPSC-CMIntraperitoneal—daily for 3d post-surgeryRats: Thoracic contusion V
In vitro reduction in LPS-induced NLRP3, CASPASE-1, IL-1β, and IL-18; reduced lesion volume; improved motor-evoked potentials and somatosensory-evoked potentials in anterior fontanelle and hind limb skeletal muscles, respectively; reduced NLRP3, IL-1β, and IL-18 in vivo; reduced microglial pyroptosis; enhanced neural repair (NF200, Tuj1, and MBP staining); reduced glial scarring (GFAP staining)Improved motor function (BBB, inclined plane test) compared to the untreated group
[106]HeP-hDPSCs, HeP-bFGF-hDSPCs10 μL of hydrogel w/ or w/o cells (no cell dose provided)Intra-spinal—immediateRats: Thoracic compression Reduced apoptotic factor expression (Bax and Caspase-3) and increased anti-apoptotic factor expression (Bcl2) in HeP-bFGF-DSPC group; increased neurogenesis (GAP43) and myelination (MBP); increased tissue and ventral motor neuron preservationGreatest motor function (BBB, inclined plane test) and sensory function (Reuters test) improvements in HeP-bFGF-DSPC and HP-DPSC groups
[107]SHED, SHED-CM1 × 106 + 1 × 105Intra-spinal fibrin glue + intrathecal pump CM—immediateRats: Thoracic contusion V
SHED and SHED-CM reduced tissue loss and spared serotonergic fibers and lesion size; SHED-CM suppressed pro-inflammatory mediators for 1 wk after injury (IL-1β and TNFα), increased expression of anti-inflammatory IL-10, TGF-β1, VEGF, CD206, and Arg-1; increased M2 macrophage phenotypes; in vitro M2 macrophage phenotype inductionSHED improved motor function (BBB) compared to the PBS control; SHED-CM improved motor function (BBB) compared to the DMEM control and BMSC-CM
[108]hDPSCs + FGF2, hDPSCs1 × 106Intra-spinal—immediateRats: Thoracic transection DPSCs and DPSC-FGF2 promoted axon regeneration (GAP-43 staining), DPSC-FGF2 more so; DPSC-FGF2 increased VEGF mRNA expressionFGF2-pretreated DPSCs significantly improved motor function (BBB) compared to the vehicle control and DPSC-only treated groups
[94]SHED3 × 105Intra-spinal—1h post-SCIRats: Thoracic contusion SHED increased neural progenitors (vimentin); SHED reduced astrocytic hypertrophy (GFAP)Improved motor function (BBB) compared to the untreated group
[109]SHED, SHED + TT3 × 105Intra-spinal—1h post-SCIRats: Thoracic contusion SHED treatment only reduced cystic cavity areas and glial–scar barrier (GFAP) caudally; SHED only increased myelin (MBP) and axonal preservation (NF-M); SHEDs only reduced intra-spinal TNFα levels (ELISA)SHED improved motor function (BBB) compared to the untreated group
[110]SHED3 × 105Intra-spinal—1h post-SCIRats: Thoracic contusion SHED treatment reduced cystic cavity areas caudally and in the lesion epicenter; motor neuron preservation and reduction in neural apoptosis; reduced T-cell infiltration and TNFα levels; reduced excitotoxic EAAT3 expressionImproved motor function (BBB) compared to the untreated group
[111]SHED2 × 105Intra-spinal—immediateRats: Thoracic compression SHED reduced p-STAT3, GFAP expression; reduced CSPGImproved motor function (BBB, inclined plane test) compared to the untreated group
[112]Rat dental pulpN/AIntra-spinal—immediateRats: Lumbar hemisection Increased motor neuron survivalNone mentioned
[113]hDPSCs (monolayer-grown), hDPSCs (spheroid-grown)3 × 105Intra-spinal fibrin glue—immediateRats: Lumbar L4-6 spinal root avulsion Increased motor neuron survival; reduced astrocyte proliferation (GFAP); reduced microglial proliferation (IBA1); preservation of neural circuitry (synatophysin); mixed inflammatory signaling changesMonolayer DPSCs improved motor function (peroneal nerve functional recovery, base of support hind paws, max contact area, and step sequence regularity)
[87]hDPSCs, SHED1 × 106 + 1 × 105Intra-spinal fibrin glue—immediateRats: Thoracic transection SHED regenerated transected corticospinal tract and seritonergic axons (DPSC not measured); SHED inhibited Rho GTPase growth inhibitor (DPSCs not measured); SHED preserved myelin sheath (Fluoromyelin and MBP) and differentiated into oligodendrocytes (DPSCs not measured); SHED reduced apoptosis of neural cells (TUNEL assay)SHED and DPSC groups improved motor function (BBB) compared to the untreated group
[114]SHED, iSHED0.5 × 106Intra-spinal—7d post-SCIRats: Thoracic contusion SHED demonstrated greater affinity for astrocytic differentiation (GFAP); iSHEDs demonstrated greater affinity for oligodendral and neural differentiation (MBP and NG2)SHED and iSHED improved motor function (BBB), more significant in the iSHED group
[115]hDPSCs, DPSC-OIC4 × 105Intra-spinal—immediateMice: Thoracic contusion V V
In vitro DPSC supernatant promoted HT-22 cell line axonal length; in vitro DPSC supernatant protected HT-22 cells from H2O2 oxidative stress-induced apoptosis; DPSC-OIC reduced hemorrhage and edema (MR imaging); DPSCs and DPSC-OIC reduced general spinal cord apoptosis (Caspase-3) and increased general cell proliferation (Ki-67); DPSCs and DPSC-OIC increased neural progenitor marker expression (Nestin) and DPSC-OIC increased progenitor marker expression (Sox2); DPSCs and DPSC-OIC reduced axon inhibitory factor NG2 and increased axon growth promoting factor fironectinDPSCs and DPSC-OIC groups improved motor function (BMS), DPSC-OIC significantly more than DPSCs only at 28d post-SCI
[116]hDFSCs, hSCAP, hDPSCs2.5 × 105Intra-spinal—immediateRats: Thoracic transection V
In vitro inhibition of general PBMC proliferation by all stem cells; promoted spinal tissue structure and neuron preservation; reduced IL-1β, RhoA, and ARHGAP growth inhibitory factors, and SUR1 necrosis and hemorrhage by all stem cells; neuronal and oligodendral differentiation (NeuN and MBP staining)All stem cell groups improved motor function (BBB) compared to the untreated group
[117]hDPSCs + TPA@laponite shear-thinning hydrogelNot provided10 μL hydrogel intra-spinal—immediateMice: Thoracic contusion Reduced lipid peroxidation (4HNE staining); increased neuronal survival closer to injury site (NeuN staining); reduced oxidation promotor expression (NOX2, GPX4, and xCT); preserved tissue integrity; reduction in ferroptosis markers; reduced fibrous blood vessel scarring and improved blood vessel organization; improved axonal regeneration (NF200 staining); regulation of excitotoxicity by reduction in Glutaminergic synapses and increase in GABAergic synapsesImproved motor function (BMS, gait mark analysis, and EMG recordings) compared to the hydrogel only and untreated groups
[118]hSCAP + ECM gel + Scramsh, hSCAP + ECM gel + MLL1sh2 × 106Intra-spinal—immediateRats: Thoracic hemisection MLL1 knockdown in SCAP reduced lesion cavities and scars than SCAP + scramsh group; increased neural progenitors (Nestin staining); increased axonal regeneration (NEFM staining);MLL1 knockdown in SCAP promoted functional recovery (BBB)
[119]hDPSCs, hDPSC + chitosan scaffold2.5 × 105Intra-spinal—7d post-SCIRats: Thoracic contusion Reduced tissue loss, apoptotic cells and axon degradation (H&E staining); reduced general apoptosis (caspase-3 expression and TUNEL staining)DPSCs and DPSC + Chitosan scaffold groups improved motor function (BBB), more so in DPSC + Chitosan scaffold group
[120]hDPSCs + GelMA hydrogel, DPSC + ZIF-8 + GelMA hydrogel0.5 × 10610 μL hydrogel intra-spinal—24 h post-SCIRats: Thoracic compression Improved tissue integrity; increased neural and blood vessel regeneration (βIII-tubulin and VEGF-α); restoration of spinal zinc levels; reduced general apoptosis (TUNEL staining)DPSCs and DPSC + ZIF8 improved motor function (BBB, inclined plan test), DPSC + ZIF8 more so, compared to the untreated group
[121]hDPSCs, AAV-5HRE-bFGF-DPSCs5 × 105Intra-spinal—7d post-SCIRats: Thoracic contusion Differentiation into pericytes, secretion of bFGF, and promotion of pericyte adhesion to vascular endothelial cells to regulate vascular diameter and reduce hypoxia; increased neuron survival and axon regeneration (NeuN and GAP43 staining); inhibited autophagy; reduced astrocytic scar (GFAP and laminin staining)DPSCs and AAV-5HRE-bFGF-DPSCs improved motor function (BBB, inclined plane test), more so in AAV-5HRE-bFGF-DPSCs group, compared to the untreated group
SHED: stem cells from human exfoliated deciduous teeth; hDPSCs: human dental pulp stem cells; hDFSCs: human dental follicle stem cells; hSCAP: stem cells from apical papilla; CM: conditioned media; BBB: Basso–Beattie–Bresnahan locomotor rating scale; BMS: Basso mouse scale; PBS: phosphate-buffered saline; HeP: heparin hydrogel; bFGF: basic fibroblast growth factor; PF-OMSF: Octyltriethoxysilane functionalized mesoporous silica (MSN) modified with PF-127 hydrogel; PF-OMSF@JK2: hydrogen sulfide gas donor JK2-loaded Octyltriethoxysilane-functionalized MSN modified with PF-127 hydrogel; Col: collagen hydrogel; HAMECs: human adipose microvascular endothelial cells; PRP: platelet-rich plasma; PAMs: pharmacologically active microcarriers; BDNF-PAMs: pharmacologically active microcarriers releasing brain-derived neurotrophic factor (BDNF); FGF2: fibroblast growth factor-2; TT: treadmill training; iSHED: neural induced SHED; OIC: Adenovirus overexpressing osteopontin, insulin-like growth factor 1, and ciliary-derived neurotrophic factor; TPA@laponite shear-thinning hydrogel: TPA(N1-(4-bor- onobenzyl)-N3-(4-boronophenyl)-N1, N1, N3, N3-tetramethylpropane-1, 3-diaminium) ROS scavenger with laponite nanoparticle hydrogel; Scramsh: scramble short hairpin RNAs; MLL1sh: MLL1 short hairpin RNAs (knockdown); GelMA: porous gelatin methacryloyl hydrogel; ZIF-8: Zn(NO3)2 nanoparticles; AAV-5HRE-bFGF: hypoxia-response element (HRE) used to mediate human bFGF with adeno-associated virus (AAV).
Table 2. Literature directly comparing the effects of dental-derived stem cells versus other mesenchymal stem cells on CNS secondary injury sequalae attenuation.
Table 2. Literature directly comparing the effects of dental-derived stem cells versus other mesenchymal stem cells on CNS secondary injury sequalae attenuation.
SCI/Non-SCIRefer-enceStem Cell TypesStudy DetailsSecondary Injury Target InvestigatedSuperior DSC Activity Compared to Other MSCs
SCI[107]SHED vs. BMSCsRat SCI contusion model; cell free CM or SHED IS engraftment; in vitro analysisNeuroinflammation; Angiogenesis; ApoptosisCM functional recovery; spinal cord M2 gene expression; in vitro CM M2 macrophage induction; VEGF secretion; neuroprotective and anti-apoptotic factor release
SCI[87]hDPSCs, SHED vs. hBMSCsRat SCI transection model; SHED IS engraftment; in vitro analysisApoptosis/Neuro-protectionNeurotrophin expression; functional recovery; in vitro neurite extension
Non-SCI[125]hDPSCs vs. hPDLSCs, hBMSCs, hAMSCsMouse palatal mucosa injury model; stem cell injectionStructural events; NeuroinflammationDPSC tissue regeneration; anti-inflammatory macrophage polarization
Non-SCI[126]hDPSCs vs. hAMSCs, hUMSCsMouse osteoporosis model; tail vein engraftmentInflammationImmunoregulatory potential of T-cell and macrophage anti-inflammatory polarization
Non-SCI[127]SHED vs. hBMSCsMouse allergic rhinitis model; IV engraftmentInflammationReduced serum IgE and IgG1 levels; decreased inflammatory cytokines in spleen; modulation of T cells
Non-SCI[128]SHED vs. hBMSCsMouse systemic lupus erythematosus model; tail vein engraftmentInflammationIncreased Treg cells to modulate inflammation
Non-SCI[129]hDPSCs vs. hBMSCsRat stroke model; hDPSC IV engraftment; in vitro ischemia analysisAngiogenesisIV engraftment efficacy; angiogenesis; in vitro neuroprotection; CM in vitro capillary formation
Non-SCI[124]SHED Mouse Alzheimer’s disease model; SHED CM intranasal administrationOxidative stress; Neuroinflammation; Neuroprotection/Anti-apoptosis3-NT reduction; in vivo anti-inflammatory environment induction; neurotrophin release
Non-SCI[130]Murine DPSCs vs. BMSCsIn vitro and in vivo naïve mouse; tibialis anterior muscle injectionAngiogenesisIn vitro vessel formation; VEGF expression; in vivo vessel formation
Non-SCI in vitro[90]hDPSCs vs. hBMSCsIn vitro trigeminal and dorsal root ganglia microfluidic assayApoptosis/Neuro-protectionNeurotrophin expression; in vitro neuronal culture axon growth
Non-SCI in vitro[123]hDPSCs vs. hBMSCs vs. hAMSCsIn vitro axotomized rat RGC analysisNeuroprotection/NeuritogenesisRGC survival vs. hAMSCs; RGC neurite extension; neurotrophin expression; VEGF expression
Non-SCI in vitro[131]hDPSCs vs. hBMSCsIn vitro neurodegeneration analysisMigrationMigration to neurodegenerative hippocampal neurons in vitro; expression of homing factors
Non-SCI in vitro[88]hDPSCs, hDFSCs, hSCAP vs. hBMSCsIn vitro neural differentiation analysisNeural differentiationNeural marker expression; CM induced neural differentiation of pre-neuroblastic cell line
Non-SCI in vitro[132]hDPSCs vs. hBMSCsIn vitro ischemia analysisOxidative stressIschemia-induced astrocyte death reduction by cells and CM
SHED: stem cells from human exfoliated deciduous teeth; BMSCs: bone marrow mesenchymal stem cells; IS: intra-spinal; CM: conditioned media; hDPSCs: human dental pulp stem cells; IV: intravenous; hAMSCs: adipose-derived mesenchymal stem cells; hUMSCs: umbilical cord-derived mesenchymal stem cells; RGC: retinal ganglion cells; hDFSCs: human dental follicle stem cells; hSCAP: human stem cells from apical papilla.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jenkner, S.; Clark, J.M.; Gronthos, S.; O’Hare Doig, R.L. Molars to Medicine: A Focused Review on the Pre-Clinical Investigation and Treatment of Secondary Degeneration following Spinal Cord Injury Using Dental Stem Cells. Cells 2024, 13, 817. https://doi.org/10.3390/cells13100817

AMA Style

Jenkner S, Clark JM, Gronthos S, O’Hare Doig RL. Molars to Medicine: A Focused Review on the Pre-Clinical Investigation and Treatment of Secondary Degeneration following Spinal Cord Injury Using Dental Stem Cells. Cells. 2024; 13(10):817. https://doi.org/10.3390/cells13100817

Chicago/Turabian Style

Jenkner, Sandra, Jillian Mary Clark, Stan Gronthos, and Ryan Louis O’Hare Doig. 2024. "Molars to Medicine: A Focused Review on the Pre-Clinical Investigation and Treatment of Secondary Degeneration following Spinal Cord Injury Using Dental Stem Cells" Cells 13, no. 10: 817. https://doi.org/10.3390/cells13100817

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