Next Article in Journal
Translational Study of Copy Number Variations in Schizophrenia
Next Article in Special Issue
Neural Crest-Derived Stem Cells (NCSCs) Obtained from Dental-Related Stem Cells (DRSCs): A Literature Review on Current Knowledge and Directions toward Translational Applications
Previous Article in Journal
Depletion of Retinal Dopaminergic Activity in a Mouse Model of Rod Dysfunction Exacerbates Experimental Autoimmune Uveoretinitis: A Role for the Gateway Reflex
Previous Article in Special Issue
Impact of Magnetic Stimulation on Periodontal Ligament Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dental Mesenchymal Stem Cell Secretome: An Intriguing Approach for Neuroprotection and Neuroregeneration

by
Agnese Gugliandolo
and
Emanuela Mazzon
*
IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(1), 456; https://doi.org/10.3390/ijms23010456
Submission received: 7 December 2021 / Revised: 28 December 2021 / Accepted: 28 December 2021 / Published: 31 December 2021

Abstract

:
Mesenchymal stem cells (MSCs) are known for their beneficial effects and regenerative potential. In particular, dental-derived MSCs have the advantage of easier accessibility and a non-invasive isolation method. Moreover, thanks to their neural crest origin, dental MSCs seem to have a more prominent neuroregenerative potential. Indeed, in basal conditions they also express neuronal markers. However, it is now well known that the beneficial actions of MSCs depend, at least in part, on their secretome, referring to all the bioactive molecules released in the conditioned medium (CM) or in extracellular vesicles (EVs). In this review we focus on the applications of the secretome derived from dental MSCs for neuroregeneration and neuroprotection. The secretomes of different dental MSCs have been tested for their effects for neuroregenerative purposes, and the secretomes of dental pulp stem cells and stem cells from human exfoliated deciduous teeth are the most studied. Both the CM and EVs obtained from dental MSCs showed that they are able to promote neurite outgrowth and neuroprotective effects. Interestingly, dental-derived MSC secretome showed stronger neuroregenerative and neuroprotective effects compared to that obtained from other MSC sources. For these reasons, the secretome obtained from dental MSCs may represent a promising approach for neuroprotective treatments.

1. Introduction

Mesenchymal stem cells (MSCs) are multipotent cells with great potential for regenerative medicine [1]. MSCs were first isolated in the bone marrow by Friedenstein et al. [2,3]. However, the term MSCs was coined later by Caplan, indicating their multipotent differentiation ability to give rise to mesodermal lineage [4]. In 2006, Dominici et al. established the criteria to classify MSCs, which are the plastic adherence ability in standard culture conditions, the expression of CD105, CD73 and CD90, the lack of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules, and the differentiation potential toward osteoblasts, adipocytes and chondroblasts in vitro [5].
Since the first discovery, MSCs have been isolated from different tissues. Regarding dental tissues, in 2000 Gronthos et al. firstly isolated a population of MSCs from dental pulp cells, with similar properties to bone marrow MSCs (BMSCs) [6]. Since then, different dental-derived cells have been found to possess stem cell properties and were named according to their tissue of origin, including dental pulp stem cells (DPSCs), stem cells from human exfoliated deciduous teeth (SHEDs), periodontal ligament stem cells (PDLSCs), dental follicle stem cells (DFSCs), stem cells from apical papilla (SCAPs), and gingival MSCs (GMSCs) [7].
Dental MSCs have the advantages of being easily accessible with minimally invasive procedures [8], expandable with relative genomic stability for a long period of time, and show immunomodulatory properties [9]. Moreover, they are also able to differentiate toward the mesodermal lineage, but they also show the ability to transdifferentiate into ectodermal and endodermal lineages [10].
Dental MSCs have neural crest origin and for this reason they show more potent neurogenic capabilities compared to other MSCs [11]. Thanks to their origin, dental MSCs express some neural progenitor and mature cell markers, even when not exposed to neural induction medium and in standard culture conditions, such as nestin, β-3 tubulin, neurotrophin receptors, and neurofilaments [12,13]. In addition, dental MSCs show a greater differentiation potential for neurogenesis compared to other MSCs types [14,15]. Thus, dental MSCs, thanks to their differentiation potential and paracrine effects, may represent a good source of MSCs for the treatment of neurodegenerative disorders and for neural regeneration [16,17,18,19,20].
The beneficial properties of MSCs are often associated with their differentiation potential. Indeed, MSCs differentiating toward neuronal cells may replace degenerated ones. However, it is now well accepted that MSCs’ regenerative and protective effects are mediated also by their secretome. In this review, we focus on the secretome obtained by dental MSCs, showing its potential for neuroprotection and neuroregeneration in preclinical models.

2. MSCs Secretome

The MSC secretome includes various bioactive molecules, such as lipids, proteins, nucleic acid, chemokines, cytokines, growth factors, and hormones, released in their conditioned medium (CM) or extracellular vesicles (EVs) [21].
The application of the secretome for cell-free therapy seems promising and has the advantage of not having the ethical limits related to the use of stem cells, and shows low immunogenicity [22]. In addition, some reports indicate only a limited survival of MSCs after transplantation [23].
EVs may also play central roles in cell-free therapies. EVs are membrane-bound bilayered lipid particles, secreted by different cell types, carrying a cargo of biological molecules from their parent cells. They are important mediators of biological information in intercellular cell signaling from the parent into a recipient cell. EVs are classified as microvesicles (MVs), exosomes (EXOs), and apoptotic bodies on the basis of their size but also of other features such as biogenesis and release pathways [24,25]. MVs are produced through direct budding from the cell plasma membrane. On the contrary, EXOs are smaller and originate by an inward budding of the limiting membrane of early endosomes, which mature into multivesicular bodies during the process. After the fusion with the plasma membrane, multivesicular bodies release EXOs into the extracellular milieu [24,26]. EVs, thanks to their surface molecules, can target the recipient cells. Once attached to a target cell, EVs can promote signaling via receptor–ligand interactions or can be internalized by endocytosis, phagocytosis or fuse with the target cell’s membrane and release their content into the cytoplasm [27,28]. EVs released by MSCs contain proteins, lipids, mRNA, microRNA (miRNA), and cytokines. These vesicles release their contents into target cells, modulating their activity and potentially inducing restorative processes [29].

Dental MSCs Secretome

Interestingly, the secretome profile may be influenced by different MSC sources [30]. For this reason, dental MSCs may present differences in secretome composition compared to other MSCs.
The analysis of SCAPs secretome has evidenced a total of 2046 proteins including chemokines, angiogenic, immunomodulatory, antiapoptotic, and neuroprotective factors other than extracellular matrix (ECM) proteins. Interestingly, the levels of 151 proteins were different by at least twofold compared to BMSCs. Indeed, SCAPs showed increased levels of proteins involved in metabolic processes, transcription, chemokines and neurotrophins while they presented a reduction of those associated with biological adhesion, developmental processes, immune function, ECM proteins and proangiogenic factors [31].
The DPSC secretome contains different cytokines, chemokines and growth factors, including vascular endothelial growth factor (VEGF)-A and Follistatin (FST) that are the most prominent [32]. Another study evidenced that granulocyte colony-stimulating factor (G-CSF)-mobilized DPSCs expressed higher levels of angiogenic and neurotrophic factors, including granulocyte macrophage colony-stimulating factor (GM-CSF), matrix metalloproteinase (MMP) 3, VEGF, and nerve growth factor (NGF) compared to BMSCs and adipose-tissue-derived MSCs (AMSCs). In particular, DPSCs-CM induced greater neurite outgrowth in human neuroblastoma TGW cells. Trophic effects of DPSCs on migration and apoptosis were higher compared to those of BMSCs and AMSCs [33]. The expression levels of cytokines in DPSCs were also compared with developing apical complex cells (DACCs). A total of 25 cytokines were identified, of which 22 were expressed more strongly in DPSCs-CM. Specifically, odontoblast differentiation-related cytokines and the neurotrophin (NT)-3 and NT-4 were more strongly expressed in DPSCs-CM [34]. The protein content of PDLSC-CM was also analyzed by liquid chromatography–tandem mass spectrometry (LC/MS/MS), that detected a total of 99 proteins, including matrix proteins, enzymes, growth factors, cytokines, and angiogenic factors [35]. LC-MS/MS also evidenced the presence of osteogenic proteins in the dental MSCs secretome [36].
Comparative secretome profiling showed the presence of fibroblast growth factor (FGF)-2, interleukin (IL)-10, platelet-derived growth factor (PDGF), stromal cell–derived factor (SDF)-1, angiopoietin (Ang)-1, transforming growth factor (TGF)-β3, hepatocyte growth factor (HGF), interferon (IFN)-γ, VEGF, and IL-6 in CM from SHEDs, BMSCs and Wharton’s-Jelly-derived MSCs (WJMSCs). PDGF-A, IL-10, FGF-2, and SDF-1 were similar in all samples, TGF-β3 and Ang-1 were higher in BMSCs, while HGF and INF-γ showed an increase in SHED. VEGF was increased in WJMSCs [37].
The differences in the secretory factors of human permanent- and deciduous-teeth PDLSCs have also been evaluated. Proteins involved in cell growth, cell communication, and signal transduction were more frequently found in permanent-teeth PDLSCs-CM, together with higher levels of NT-3 and NT-4 and angiogenesis-related cytokines such as epidermal growth factor (EGF) and insulin-like growth factor (IGF)-1. On the contrary, CM obtained from deciduous-teeth PDLSCs contained mainly proteins involved in regulation of the cell cycle and the levels of cytokines involved in immune response and in tissue degradation and catalytic activities, including MMP1, Proteasome subunit, alpha type, 1 (PSMA1), and cullin 7 (CUL7) were higher in these cells [38].
Pulp CD31 side population (SP) cells expressed the highest levels of angiogenic and neurotrophic factors compared to those isolated from bone marrow and adipose tissue. CM from pulp CD31 SP cells showed anti-apoptotic and neurite outgrowth ability [39].
EXOs derived from DPSCs showed stronger immune-modulating capacity compared to BMSCs EXOs. Specifically, DPSCs EXOs inhibited CD4+ T cell differentiation into T helper 17 cells and reduced the secretion of pro-inflammatory cytokines IL-17 and tumor necrosis factor (TNF)-α, while promoting the polarization of CD4+ T cells into T reg and increasing the release of the anti-inflammatory factors IL-10 and TGF-β [40].
The transcripts present in EVs were studied also. GMSCs EVs contained transcripts encoding for several growth factors such as TGF-β, FGF, and VEGF, but also glial-cell-derived neurotrophic factor (GDNF) family ligands and neurotrophins, such as NGF, brain-derived neurotrophic factor (BDNF), NT-3 and NT-4 involved in neuronal development. Some ILs and members of the Wnt family were also present [41].
EVs also contain non-coding RNA. EVs of PDLSCs highlighted the presence of different classes of non-coding RNAs, including antisense RNA and long non-coding RNA (lncRNAs), but also five miRNAs, which are MIR24-2, MIR142, MIR335, MIR490, and MIR296. These miRNAs target genes belonged to the gene ontology class “Ras protein signal transduction” and “Actin/microtubule cytoskeleton organization” [42].
A total of 593 and 920 known PIWI-interacting RNAs (piRNAs) were identified from SCAP-EXOs and BMSC-EXOs, respectively, and 21 piRNAs were differentially expressed. The target genes of the differentially expressed piRNAs were mainly involved in biological regulation, cellular processes, metabolic processes, binding, and catalytic activity. Specifically, the target genes of the upregulated piRNAs in SCAP-EXOs were enriched in the mitogen-activated protein kinase (MAPK) signaling pathway, Ras signaling pathway, and citrate cycle signaling pathway. On the contrary, the target genes of downregulated piRNAs in SCAP-EXOs were enriched in the p53 signaling pathway and Epstein–Barr virus infection signaling pathway [43].
It is important to notice that donor age and microenvironmental conditions in vitro may also influence secretome composition. DPSC-CM obtained in normoxic conditions was reported to be enriched in molecules with anti-inflammatory, tissue repair, and regenerative properties compared to CM obtained in hypoxic conditions [44]. Moreover, secretome collected from 5% O2 cultured DPSCs showed higher stimulatory effects on proliferation and migration of mouse embryonic fibroblast NIH3T3 cells and on neuronal differentiation of SH-SY5Y cells [45]. The quantity and size of EXOs and their tetraspanins expression may vary dependent on the medium used for culture [46]. The secretome of SHEDs and young DPSCs contained more growth factors and lower levels of pro-inflammatory cytokines compared to DPSCs obtained from old subjects. Differentiation potential was also higher in SHEDs and young DPSCs [47].
CM can be obtained by healthy PDLSCs but also by inflamed PDLSCs. CM obtained by inflamed ones increased proliferation of both healthy and inflamed PDLSCs, but reduced the differentiation toward osteoblasts. Healthy CM rescued the impaired osteogenic differentiation [48].
The treatment with different substances may also influence cell secretome. The treatment of the DPSCs with 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside (THSG), a bioactive component of Polygonum multiflorum Thunb., induced changes in the secretion of growth-associated proteins in CM, increasing some of them such as AKT2 and NGF receptor [49]. Instead, CM from FGF-2-modified GMSCs contained more VEGF-A, FGF-2, and TGF-β [50]. Ascorbic acid treatment of SHEDs increased the release of growth factors necessary for the tissue regeneration and homeostasis, including VEGF, SCF, IGF-1, HGF, bFGF, Ang-1, and EGF, and anti-inflammatory cytokines, such as NO, indoleamine 2,3-dioxygenase (IDO), PGE-2, IL-10, and IL-6. On the contrary, inflammatory cytokines CCL2 and TGF-β1 were reduced [51].
The exposure to differentiation medium also could induce changes in non-coding RNA in EVs and EXOs of PDLSCs. Specifically, 69–557 circular RNA (circRNAs) and 2907–11,581 lncRNAs were found in EVs isolated from PDLSCs and PDLSCs exposed to osteogenic differentiation medium at different time points. Compared with undifferentiated PDLSCs EVs, 3 circRNAs and 2 lncRNAs were upregulated and 39 circRNAs and 5 lncRNAs were downregulated consistently after 5 and 7 days of exposure to differentiation medium [52]. Moreover, 72 miRNAs were upregulated while 35 were downregulated in PDLSCs EXOs after osteogenic induction [53].
A summary of the main factors found in the secretome of the different dental MSCs can be found in Table 1.

3. Dental Stem Cell Secretome Neuroprotective and Neuroregenerative Potential in Preclinical Models

In order to evaluate the neuroregenerative and neuroprotective potential of the dental MSC secretome, the effects of CM and EVs have been evaluated in preclinical models of neurodegenerative and neurological diseases and models of neuronal damage, such as spinal cord injury (SCI). In addition, secretome mediated effects on neuronal outgrowth, its capacity to stimulate neuronal differentiation, and the effects on glial cells have also been evaluated. We performed a PubMed search looking for studies showing the neuroregenerative and neuroprotective potential of dental MSCs secretome in in vitro and in vivo models.

3.1. Dental Pulp Stem Cell Secretome

DPSCs secretome was one of the most studied. Different studies evaluated its efficacy in inducing neurite outgrowth. It was reported that DPSC-CM promoted neurite outgrowth in dorsal root ganglion (DRG) neurons. Specifically, the total length and joint number of neurites increased after treatment with CM. Moreover, DPSC-CM promotes Schwann cell viability and myelin formation [54].
DPSCs-CM enhanced cell survival and induced neurite outgrowth of PC12 cells, as shown by neuronal nuclear protein (NeuN), microtubule-associated protein 2 (MAP-2) and βIII-tubulin. Specifically, DPSCs-CM was more efficacious in inducing PC12 neurite outgrowth compared to DPSCs/PC12 co-cultures, indicating that cells co-cultures had a delayed lag time in producing efficacious amounts of trophic factors. DPSCs-CM also enhanced cell migration. Interestingly, the number of surviving PC12 cells was reduced when CM was added with anti-GDNF. Instead, the addition of anti-NGF, anti-GDNF and anti-BDNF antibodies attenuate PC12 neurite outgrowth. These data demonstrated that NGF, BDNF and GDNF are involved in the PC12 survival and differentiation [55].
The DPSC secretome shows a chemoattractive effect on SH-SY5Y cells. Moreover, its effect on neural maturation has been evaluated. With this aim, SH-SY5Y cells were induced toward neuronal cells and after they were exposed to the DPSC secretome. SH-SY5Y cells subjected to the DPSC secretome showed increased neurite outgrowth, acquired ultrastructural features of neuronal cells and presented an increased immune reactivity for neuronal markers. Moreover, CM-treated SH-SY5Y cells developed distinct features including Cd2+-sensitive currents, which suggests that CM-DPSC-maturated SH-SY5Y acquired voltage-gated Ca2+ channels [56]. In line with the previous study, CM obtained by DPSC sheet induced the formation and outgrowth of neurites in neuronally differentiated SH-SY5Y neuroblastoma cells. These effects were enhanced when DPSC sheets were cultured with FGF2. The neurite-promoting effects were abolished when neurotrophic factors were inhibited, suggesting that they are needed for the positive effect of DPSC sheets on neuronal cell activity [57].
Recently, Chouaib et al. evidenced that DPSC-CM enhancement of neurite outgrowth in sensory neurons is concentration dependent. The authors also found that 48 h of DPSCs conditioning was the best option to obtain CM with efficient activity, while extending the conditioning time did not improve the effects of DPSC-CM. Interestingly, the frozen storage did not influence experimental outcomes. The CM contained some factors known for their role in neurogenesis and neuroprotection but also in angiogenesis and osteogenesis. Moreover, the conditioning of DPSCs with the B-27 supplement enhanced the neuroregenerative effects of their secretome, inducing a change of its composition in growth factors. In particular, CM was more efficacious when B-27 was added to DPSCs before conditioning [58].
CM from DPSCs enhanced neuritogenesis and exerted a chemoattractant effect also on neural stem cells (NSCs). The priming of DPSCs with leukocyte- and platelet-rich fibrin (L-PRF) increased BDNF secretion, but exerted no additional effects on the paracrine-mediated repair mechanisms [59].
DPSC-derived CM was also shown to be able to protect and regenerate isolated primary trigeminal ganglion neuronal cells (TGNC). Indeed, CM enhanced TGNC survival associated with extensive neurite outgrowth and branching. In parallel, DPSC-CM significantly upregulated NeuN, βIII-tubulin, and synapsin-I neuronal marker expression as well as TRPV1. Interestingly, DPSC-CM contained NGF, BDNF, NT-3, and GDNF [60].
G-CSF-mobilized DPSCs expressed higher neurotrophic factors compared to basal DPSCs and their secretome showed an enhanced neurite extension potential. Indeed, mobilized DPSC CM had a greater effect on neurite outgrowth in TGW cells [61]. Previously, it was demonstrated that CM from mobilized DPSC enhanced proliferation and migratory activity of neuronal Schwann RT4-D6P2T cells [62].
Interestingly, CM from SHEDs and DPSCs was shown to be able to promote the regeneration of cerebral granular neurons inhibiting axon growth inhibitors signals by paracrine mechanisms [63].
The DPSCs secretome also shows superior effects compared to other MSCs. Kumar et al. demonstrated that the secretome derived from DPSCs, SCAPs and DFSCs induced neural differentiation in IMR-32 cells, a preneuroblastic cell line, in a more efficient manner compared to BMSCs. In particular, neurite length was higher when IMR-32 cells were treated with the DPSC secretome. The DPSC secretome contained GCSF, IFN-γ, and TGF-β, which may promote neural differentiation [64].
DPSCs, BMSCs and AMSCs promoted an increase in the survival of co-cultured retinal ganglion cells. In particular, the increase in survival was enhanced in DPSC-treated retinal cultures. Interestingly, coculture with DPSC induced a significant increase in both the number of neurite-bearing retinal ganglion cells and neurite length compared with cocultures with BMSCs and AMSCs. However, these effects were blocked using neurotrophic factor receptors Fc-receptor blockers. The different types of MSCs showed a different pattern of neurotrophic factor expression, and, specifically, DPSCs released higher levels of several growth factors such as NGF, BDNF, and VEGF compared with BMSCs and AMSCs. In particular, VGF may mediate the neuroprotective effects of DPSCs [65].
CM-DPSCs showed protective effects on oxygen-glucose deprivation (OGD)-induced cytotoxicity in astrocytes in a dose-dependent manner. Specifically, both pre- and posttreatment with CM-DPSCs, but also CM-BMSCs, attenuated OGD-induced glial fibrillary acidic protein (GFAP), nestin, and musashi-1 expression in astrocytes. The treatment with CM also blocked OGD-induced reactive oxygen species (ROS) production and IL-1β upregulation. Interestingly, CM-DPSCs confer superior cytoprotection against cell death compared with BMSCs [66].
Venugopal et al. compared the neuroprotective potential of EXOs, CM or neuron–MSC-co-culture system against kainic-acid-induced excitotoxicity in vitro. Moreover, in order to identify the most adapt MSC type, EXOs and CM derived from DPSCs and BMSCs were tested. All three approaches showed neuroprotective potential thanks to the increase of growth factor expressions and the inhibition of apoptosis through the activation of PI3K-Bcl-2 pathway. It is important to note that EXOs demonstrated better anti-necrotic properties compared to neuron-MSC co-culture or CM. Regarding CM, only the fraction containing proteins in the range 3–10 kDa showed neuroprotection and rescued the neurons from excitotoxicity [67].
The secretome of DPSCs also showed beneficial effects in models of neurodegenerative diseases. Treatment with DPSC secretome reduced amyloid β (Aβ) cytotoxicity in an in vitro model of Alzheimer’s disease (AD), increasing cell viability and reducing apoptosis. DPSC secretome was shown to contain elevated levels of VEGF, Fractalkine, RANTES, monocyte chemoattractant protein-1 (MCP-1), and GMCSF compared to those of BMSCs and AMSCs. Interestingly, neprilysin, a protease able to degrade Aβ, was also found in the DPSC secretome. DPSC secretome proteolytically degrades Aβ1–42 in vitro, resulting in complete degradation after 12 h [68].
Early pre-symptomatic DPSC-CM administration improved neuromuscular junction innervation compared to vehicle-treated SOD1G93A mice. The administration during late pre-symptomatic stages not only increased neuromuscular junction preservation, but also motor neuron survival in the spinal cord ventral horn. However, astrogliosis and microglia reactivity remained unaffected. Interestingly, the daily DPSC-CM treatment from symptom onset increased post-onset survival as well as overall lifespan [69].
DPSC-EXOs administration in a murine model of transient middle cerebral artery occlusion (tMCAO) injury reduced brain oedema, cerebral infarction, and neurological impairment. DPSC-EXOs inhibited the ischemia/reperfusion (I/R)-mediated expression of Toll-like receptor (TLR) 4, MyD88, and Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB). DPSC-EXOs also reduced the protein expression of pro-inflammatory cytokines IL-6, IL-1β and TNF-α, and cytoplasmic translocation of high mobility group box protein (HMGB) 1 in vivo but also in vitro in OGD/reperfusion (OGD/R)-induced BV2 cells. Thus, the results indicated that DPSC-EXOs may exert neuroprotection against cerebral I/R-induced neuroinflammation via the inhibition of the HMGB1/TLR4/MyD88/NF-κB signaling pathway [70].
DPSC-CM ameliorated aneurysmal subarachnoid hemorrhage (aSAH)-induced vasoconstriction and improved oxygenation in an injured brain. DPSC-CM administration also ameliorated cognitive and motor impairments. DPSC-CM administration decreased neuroinflammation as demonstrated by the reduction in the number of Iba1-positive cells. The major constituent of DPSC-CM was IGF-1. Antibody-mediated neutralization of IGF-1 moderately deteriorated the rescuing effect of DPSC-CM on microcirculation, Iba1-positive cells in the injured brain area, and the cognitive/motor impairments [71].
DPSC-CM administration ameliorated sciatic motor/sensory nerve conduction velocity, sciatic nerve blood flow, and intraepidermal nerve fiber density in the footpads of streptozotocin-induced diabetic rats. Furthermore, capillary density of the skeletal muscles increased while pro-inflammatory reactions in the sciatic nerves of diabetic rats were reduced [72]. Kanada et al. confirmed the positive effects of CM in sciatic nerve conduction velocity and sciatic nerve blood flow. Moreover, the treatment also increased muscle bundle size, vascular density in the skeletal muscles, and intraepidermal nerve fiber density in the diabetic rats. However, no differences were found between the results for DPSCs and DPSC-CM. These results suggested that the efficacy of DPSC and DPSC-CM administration were probably due to the secretome. In particular, DPSC-CM contained angiogenic factors such as VEGF-C, neurotrophic factors, such as BDNF, and immunomodulatory factors including IL-1β, IL-4, and TLR4 [73].
An overview of the studies presented in this paragraph is available in Table 2.

3.2. Stem Cells from Human Exfoliated Deciduous Teeth Secretome

Secretome of SHEDs was reported to modulate microglial cell activity. EVs derived from SHEDs inhibited lipopolysaccharide (LPS)-induced activation of NF-κB signaling pathway in human microglial cells. Moreover, EVs induced an upregulation of phagocytic activity in unpolarized cells, a slight decrease in M1 polarized cells, and a moderate increase in M2 polarized cells. EVs induced an immediate and sustained increase of glycolytic activity in M0, M1, and M2 polarized cells. Interestingly, EVs acted in an inverse dose-dependent manner [74]. EVs also induced a rapid increase in intracellular Ca2+ and ATP release in microglial cells. EVs were also able to promote microglial motility through P2X4 receptor/milk fat globule-epidermal growth factor-factor VIII (MFG-E8)-dependent mechanisms [75].
Different studies report beneficial effects of SHED CM in both in vitro and in vivo Parkinson’s disease (PD) models. Fujii et al. evidenced that dopaminergic-neuron-like cells induced from SHEDs were able to exert therapeutic benefits in a 6-hydroxy-dopamine (6-OHDA)-induced Parkinsonian rat model, improving neurological deficits and increasing dopamine (DA) levels more efficiently than undifferentiated SHEDs. However, paracrine effects may contribute to neuroprotection against 6-OHDA-induced neurodegeneration. Indeed, the CM obtained from differentiated SHEDs was able to protect primary neurons against 6-OHDA toxicity and accelerated neurite outgrowth in vitro [76]. Different doses of SHED-CM were tested in a PD model. The dose 10 µg/mL of SHED-CM did not restore motor ability, while 30 µg/mL of SHED-CM induced only mild improvements. Instead, 100 µg/mL of SHED-CM induced the maximal improvement of motor deficits in PD rats and a higher dose did not induce further improvement. SHED-CM increased tyrosine hydroxylase (TH) amounts and decreased synuclein levels in both the substantia nigra and striatum. In addition, SHED-CM treatment decreased both Iba-1 positive cells and CD4 levels in the same brain areas. The major constituents of SHED-CM included insulin-like growth factor binding protein-6 (IGFBP-6), tissue inhibitor of metalloproteinase (TIMP)-2, TIMP-1, and TGF-1. Moreover, bioinformatics analysis indicated that SHED-CM was able to promote neural regeneration. Indeed, RNA sequencing evidenced that SHED-CM administration shifted the gene expression profile to a pattern similar to that of control rats, upregulating genes that were involved in neurodevelopment and nerve regeneration. The major constituents of SHED-CM may participate in the molecular networks involved in cholinergic and serotoninergic synapses, calcium signaling pathways, and axon guidance [77].
EXOs and MVs derived from SHEDs have also been evaluated for their neuroprotective effects in PD models. EXOs, but not MVs, derived from SHEDs grown on laminin-coated three-dimensional alginate micro-carriers suppressed 6-OHDA-induced apoptosis in dopaminergic neurons. On the contrary, no protective effects were exerted by MVs or EXOs derived from SHEDs grown in standard culture conditions [78].
Instead, intranasal administration of EVs derived from SHEDs was shown to be effective in a rat model of PD, improving motor function in association with a normalization of TH expression in the striatum and substantia nigra [79].
The intranasal administration was also tested in an AD model, showing that SHED-CM improved cognitive function. SHED-CM reduced oxidative stress, shifted the M1-type pro-inflammatory microenvironment toward the M2-type anti-inflammatory and neuroprotective one, and increased neurotrophic factor levels. BMSCs-CM was less efficacious. It reduced oxidative stress and inflammation, but could not upregulate the expression of the anti-inflammatory M2 markers. Treatment with SHED-CM also suppressed glutamate-induced neuronal death in vitro [80].
SHED-CM was also able to improve disease scores and reduce demyelination, axonal injury, inflammatory cell infiltration, and proinflammatory cytokine expression in the spinal cord of experimental autoimmune encephalomyelitis (EAE) mice. These changes were associated with a change in the microglia/macrophage phenotype from M1 to M2. The treatment of EAE mice with the secreted ectodomain of sialic-acid-binding Ig-like lectin-9 (ED-Siglec-9), a major component of SHED-CM, resulted in similar effects compared to SHED-CM treatment, while ED-Siglec-9 depletion abolished the protective effects of SHED-CM. On the contrary, HGF depletion did not cause an inhibition of SHED-CM mediated protection, indicating that HGF had little effect on the efficacy of SHED-CM. SHED-CM inhibited the proliferation of myelin oligodendrocyte glycoprotein-specific CD4+ T cells, as well as their production of proinflammatory cytokines in vitro [81].
Matsubara et al. showed that SHED and SHED-CM administered into rat injured spinal cord during the acute postinjury period induced functional recovery. SHED-CM showed anti-inflammatory activity, reducing the levels of pro-inflammatory cytokines, and immunoregulatory action, inducing M2 anti-inflammatory macrophages. To identify factors responsible for the therapeutic effects of CMs, soluble factors present in SHED-CM were characterized. A total of 79 proteins were identified, some of them known to be involved in neuroregenerative processes, with anti-apoptotic, anti-inflammatory and axonal elongation properties. In particular, MCP-1 and ED-Siglec-9 may be involved in M2-like macrophage differentiation. Indeed, depleting these factors from the SHED-CM reduced CM’s ability to induce M2-like macrophages and to promote functional recovery after SCI. Interestingly, the administration of BMSC-CM induced no or only slight M2-like cell differentiation and did not induce recovery such as SHED-CM [82].
In agreement with the previous study, the treatment with SHED-CM loaded on a collagen hydrogel, used as a delivery system, induced functional recovery in SCI rats, as demonstrated by improvement in scores evaluated through Basso, Beattie, and Bresnahan scoring, inclined plane, cold allodynia, and beam walk tests [83]. The treatment with SHED-CM loaded on a collagen hydrogel also increased the volume of preserved white and gray matter and the total number of neurons and oligodendrocytes in a rat SCI model. On the contrary, lesion volume and lesion length decreased. However, in this study SHED-CM alone exerted no protection. The authors suggested that this may be due to the rapid diffusion of SHED-CM, and thus collagen hydrogel may act as an efficient releasing system [84].
A single intravenous injection of SHED-CM also reversed the mechanical allodynia induced by spinal nerve transection, suppressed microglia and astrocytes activation, and decreased the numbers of neurons positive for the neuronal injury marker activating transcription factor 3 (ATF3) and macrophage accumulation. In particular, the SHED-CM fraction with a molecular weight between 30 and 50 kDa reversed the pain, suggesting that protein components with molecular mass in the range 30–50 kDa were responsible for the reported neuroprotection [85].
The implantation of a collagen sponge enriched with the serum-free CM from SHED into the nerve gap formed by rat facial nerves transection restored the neurological function. On the contrary, CM depleted of MCP-1 and ED-Siglec-9, which are anti-inflammatory M2 macrophage inducers, did not restore neurological function. Notably, MCP-1 and ED-Siglec-9 induced the polarization of M2 macrophages in vitro and in vivo. Thus, the results indicated that MCP-1/ED-Siglec-9 participated in peripheral nerve regeneration inducing M2 macrophage [86].
SHED-CM treatment increased proliferation, migration, and the expression of neuron-, ECM-, and angiogenesis-related genes in Schwann cells. Moreover, SHED-CM stimulated neurite outgrowth of dorsal root ganglia and increased cell viability. In vivo, axon regeneration and myelination were higher in the SHED-CM group after nerve transection surgery. Motor function improved while muscle atrophy was reduced in the SHED-CM group. Thus, SHEDs may secrete various trophic factors that enhance peripheral nerve regeneration through multiple mechanisms. Specifically, SHED-CM contained NGF, BDNF, NT-3, GDNF, ciliary neurotrophic factor (CNTF), VEGF, and HGF [87].
The administration of SHED-EXOs improved rat motor functional recovery and reduced cortical lesion in rats with traumatic brain injury. SHED-EXOs can exert these effects, reducing neuroinflammation shifting microglia polarization [88].
SHED-CM induced an improvement of motor disability and reduced infarct volume after permanent MCAO. The SHED-CM treated group showed increased levels of doublecortin, neurofilament H, neuronal nuclei and rat endothelial cell antigen in the peri-infarct area. Interestingly, SHED-CM induced the migration and differentiation of endogenous neuronal progenitor cells (NPC), vasculogenesis and improved ischemic brain injury [89].
Intracerebral administration of SHED-CM in hypoxia-ischemia injured mice improved neurological function, survival rate, and neuropathological score [90].
CM obtained from SHEDs, and specifically only the fraction of <6 kDa, promoted neurite outgrowth of DRG neurons. Moreover, SHED-CM prevented the decline in sensory nerve conduction velocities in diabetic mice and ameliorated the capillary number-to-muscle fiber ratio and capillary blood flow [91].
In an animal model of superior laryngeal nerve injury, the systemic administration of SHED-CM induced functional recovery, increasing the degree of myelination and promoted axonal regeneration shifting macrophages toward the M2 phenotype [92].
An overview of the studies presented in this paragraph is available in Table 3.

3.3. Periodontal Ligament Stem Cell Secretome

CM obtained from PDLSCs of relapsing remitting multiple sclerosis (RR-MS) patients showed anti-inflammatory and antiapoptotic effects in NSC-34 mouse motor neurons stimulated with the medium of LPS treated RAW 264.7 macrophages. Indeed, CM treatment reduced TLR4 and NF-κB levels, together with pro-inflammatory cytokines. On the contrary, IL-10 increased in association with neuroprotective markers such as nestin, neurofilament 70, NGF, and GAP43. Interestingly, the neuroprotective effects of EVs may be due to their content in IL-10 and TGF-β [93].
The effects of PDLSCs-CM obtained from healthy donors and RR-MS patients were also evaluated in phorbol-12-myristate-13-acetate (PMA) differentiated THP-1, used as a model of microglia, and in undifferentiated and differentiated MO3.13 cells, used as models of progenitor cells and oligodendrocytes, respectively, treated with Porphyromonas gingivalis LPS. Treatment with both CM reduced the LPS-induced increase in TNFα, IL-1β and IL-6 levels and reduced TLR-4 in THP-1 cells [94].
PDLSCs-CM and purified EXOs/MVs (PDLSCs-EMVs) obtained from RR-MS patients and healthy donors exerted protective effects in EAE mice. In particular, PDLSCs-CM and PDLSCs-EMVs improved disease scores, restoring tissue integrity and remyelination in the spinal cord. PDLSCs-CM and PDLSCs-EMVs exerted anti-inflammatory effects both in spinal cord and spleen, as demonstrated by the reduction of pro-inflammatory cytokines and the induction of IL-10. In parallel, apoptosis was also inhibited. The anti-inflammatory effect of CM or EMVs might be due to the presence of the immunomodulatory cytokines IL-10 and TGF-β [95]. Moreover, PDLSCs-CM and EMVs obtained from RR-MS patients inhibited NALP3 inflammasome activation and reduced TLR-4 and NF-κB levels in EAE mice. The immunomodulatory factors IL-10, TGF-β, and SDF-1α included in the CM may be responsible for the immunosuppressive role of PDLSCs-CM and EMVs in EAE [96].
Interestingly, CM obtained from PDLSCs cultured under hypoxic conditions was efficacious in ameliorating clinical and histologic disease scores in EAE mice. In particular, this treatment reduced inflammatory cell infiltration and increased remyelination in the spinal cord. In particular, hypoxic CM administration increased the anti-inflammatory cytokine IL-37 in association with the reduction of proinflammatory cytokines. Moreover, oxidative stress and apoptosis were also inhibited, while BDNF increased. CM treatment was also able to regulate autophagy through the activation of the PI3K/Akt/mTOR pathway. Furthermore, in in vitro scratch injury exposed NSC-34 motor neurons, the hypoxia CM was able to modulate inflammation, oxidative stress, and apoptosis. Interestingly, hypoxia CM contained NT-3, IL-10, and TGF-β that may explain its neuroprotective effects [97].
An overview of the studies presented in this paragraph is available in Table 4.

3.4. Other Dental-Derived MSCs

Neuroprotective effects of CM obtained by GMSCs were evaluated in mechanically injured murine motor-neuron-like NSC-34 cells. CM treatment reduced the scratch injury-induced apoptosis and oxidative stress. Moreover, CM reduced TNF-α while increasing the levels of the anti-inflammatory cytokine IL-10. Interestingly, CM treatment upregulated BDNF and NT-3. CM was shown to contain NGF, NT-3, IL-10, and TGF-β, which may explain the neuroprotective effects [98].
EVs from GMSCs were tested for peripheral nerve regeneration in a crush-injured sciatic nerve mouse model. In vivo, the transplantation of Gelfoam embedded with GMSC-derived EVs at the crush injury site induced functional recovery and axonal regeneration in a similar way compared to the direct transplantation of GMSCs. In particular, EVs promoted proliferation and migration of Schwann cells and upregulated the protein expressions of c-JUN, Notch1, GFAP, and SOX2 genes associated with dedifferentiation or repair phenotype of Schwann cells. Also in vitro, EVs promoted the expression of Schwann cell dedifferentiation/repair genes [99]. A positive effect on Schwann cell proliferation was also reported for EXOs from GMSCs, which also promoted DRG axon growth in vitro. Moreover, the effects of GMSCs EXOs combined with biodegradable chitin conduits on peripheral nerve regeneration were evaluated. In vivo, in a rat sciatic nerve defect model, chitin conduit combined with EXOs increased the number and diameter of nerve fibers and promoted myelin formation. In parallel, nerve conduction also improved. Moreover, muscle function and motor function were ameliorated [100].
CM from SCAPs, DPSCs, and PDLSCs were tested to evaluate their capacity in inducing neurite outgrowth. With this aim, differentiated neuroblastoma SH-SY5Y cells were incubated with the different CM. The CM were shown to be able to increase the percentage of cells producing neurites and the total neurite outgrowth length. Interestingly, the length of the longest neurite per neuron was increased in a significant manner only with SCAP CM, and the neutralization of the secreted BDNF inhibited neurite outgrowth, indicating its importance in this process [101]. CM released from SCAPs showed also a greater neurogenic inductive effect on DPSCs compared to BMSCs-CM. Indeed, when DPSCs were cultured in medium for neural stem cell growth, the levels of neurogenic markers increased with the addition of SCAPs-CM. On the contrary, neuronal marker expression was reduced, while that of neurotrophic marker increased, when BMSCs-CM was added. Cell proliferation was not influenced by SCAPs-CM [102].
Oral mucosa stem cells (OMSCs) were differentiated into cells showing an astrocyte-like morphology and expressed characteristic astrocyte markers. The CM obtained by differentiated OMSCs increased the cell viability of motor neurons cultured in hypoxic conditions or exposed to hydrogen peroxide in vitro [103].
An overview of the studies presented in this paragraph is available in Table 5.

4. Translation of Secretome Application from the Preclinical Models into Clinical Use

The application of the secretome for a cell-free therapy may present some advantages compared to the use of MSCs. The main advantages of the use of the secretome instead of stem cell therapy are represented by the low immunogenicity and easier production, handling, and storage of the secretome [104]. Then, this therapy can overcome the risks linked to cell therapy, such as tumorgenicity, antigenicity, host rejection, and infections. Secretome handling can be easier compared to cells, given that it can be concentrated, frozen, and that it does not require liquid nitrogen storage and cell culture facilities, making its transfer also easier [105,106]. Moreover, secretome production is more economical and mass production under controlled laboratory conditions is also possible [107]. However, the period of survival of MSCs after transplantation is not clear, given that some data seem to indicate a limited survival [23].
Thus, therapeutic strategies based on secretome application could be helpful in regenerative medicine, based on their content in bioactive molecules, including proteins, mRNAs but also non-coding RNA, that can be useful to induce repair mechanism in the injured tissues. However, before translation into clinical applications, several points need to be clarified. In particular, it is necessary to better define the secretome composition, dosage, frequency, and route of administration. In this regard, it is also necessary to develop standardized manufacturing protocols with good manufacturing practice for the development of new pharmaceuticals based on cell-free products [105]. Indeed, for applications into clinical practice, the secretome should be presented in a standardized and easy to handle formulation. The secretome indeed may present variations on the basis of the subjects, cells, and tissues of origin [30]. For this reason, to standardize the secretome production it is necessary to define culture medium and supplements, culture duration, and culture conditions [106]. In this context, big data studies that evaluate proteome, transcriptome and non-coding RNA profile may provide assistance for secretome characterization. A list of studies analyzing transcriptome, non-coding RNA, and proteome profiling is available in Table 6.
However, secretome delivery also needs to address the concern of the rapid diffusion and clearance of secretome from the tissue repair site. Moreover, secretome stability and the stability of its growth factors and miRNAs need to be maintained in physiological conditions for all the delivery duration. In this context, different biomaterials have been developed and optimized in order to improve the delivery efficiency of MSCs secretome, with the advantages of a prolonged release duration, protection against degradation, and enhancing the therapeutic capacity [108]. Another option to improve the long-lasting stability of formulations is a freeze-drying process, which is used for different biological products [109,110].

5. Conclusions

Dental MSCs, given their origin from neural crest, have been shown to possess a prominent neuroregenerative potential. Dental MSC-derived secretome also shows the same enhanced neuroprotective and neuroregenerative properties. Both CM and EVs contain neurotrophins and molecules with a neuroprotective action, even at higher levels compared to other MSCs. The studies evaluated in this review highlighted that both CM and EVs stimulated neurite outgrowth and showed neuroprotective effects in preclinical models of neurological diseases and neuronal damage. In particular, the secretomes of DPSCs and SHEDs were the most studied, but different studies also highlighted the neuroprotective effects of PDLSC and GMSC secretomes. Interestingly, some studies also suggested the superiority of the secretome obtained from dental MSCs compared to other MSCs sources, such as BMSCs and AMSCs, for neuroprotection. In conclusion, the secretome derived from dental MSCs seems promising for its application in the neuroregenerative field and may be useful to develop new neuroprotective treatments.

Author Contributions

Conceptualization, E.M.; writing—original draft preparation, A.G.; writing—review and editing, E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Current Research Funds 2021, Ministry of Health, Italy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funder had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Vasanthan, J.; Gurusamy, N.; Rajasingh, S.; Sigamani, V.; Kirankumar, S.; Thomas, E.L.; Rajasingh, J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020, 10, 54. [Google Scholar] [CrossRef] [PubMed]
  2. Friedenstein, A.J.; Petrakova, K.V.; Kurolesova, A.I.; Frolova, G.P. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968, 6, 230–247. [Google Scholar] [CrossRef] [PubMed]
  3. Friedenstein, A.J.; Chailakhjan, R.K.; Lalykina, K.S. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970, 3, 393–403. [Google Scholar] [CrossRef]
  4. Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. Off. Publ. Orthop. Res. Soc. 1991, 9, 641–650. [Google Scholar] [CrossRef]
  5. 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]
  6. 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] [PubMed] [Green Version]
  7. Li, B.; Ouchi, T.; Cao, Y.; Zhao, Z.; Men, Y. Dental-Derived Mesenchymal Stem Cells: State of the Art. Front. Cell Dev. Biol. 2021, 9, 654559. [Google Scholar] [CrossRef]
  8. Egusa, H.; Sonoyama, W.; Nishimura, M.; Atsuta, I.; Akiyama, K. Stem cells in dentistry-part I: Stem cell sources. J. Prosthodont. Res. 2012, 56, 151–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Gan, L.; Liu, Y.; Cui, D.; Pan, Y.; Zheng, L.; Wan, M. Dental Tissue-Derived Human Mesenchymal Stem Cells and Their Potential in Therapeutic Application. Stem Cells Int. 2020, 2020, 8864572. [Google Scholar] [CrossRef]
  10. Yin, J.Y.; Luo, X.H.; Feng, W.Q.; Miao, S.H.; Ning, T.T.; Lei, Q.; Jiang, T.; Ma, D.D. Multidifferentiation potential of dental-derived stem cells. World J. Stem Cells 2021, 13, 342–365. [Google Scholar] [CrossRef]
  11. Mayo, V.; Sawatari, Y.; Huang, C.Y.; Garcia-Godoy, F. Neural crest-derived dental stem cells--where we are and where we are going. J. Dent. 2014, 42, 1043–1051. [Google Scholar] [CrossRef]
  12. Luzuriaga, J.; Polo, Y.; Pastor-Alonso, O.; Pardo-Rodriguez, B.; Larranaga, A.; Unda, F.; Sarasua, J.R.; Pineda, J.R.; Ibarretxe, G. Advances and Perspectives in Dental Pulp Stem Cell Based Neuroregeneration Therapies. Int. J. Mol. Sci. 2021, 22, 3546. [Google Scholar] [CrossRef]
  13. Heng, B.C.; Lim, L.W.; Wu, W.; Zhang, C. An Overview of Protocols for the Neural Induction of Dental and Oral Stem Cells In Vitro. Tissue Eng. Part B Rev. 2016, 22, 220–250. [Google Scholar] [CrossRef]
  14. Isobe, Y.; Koyama, N.; Nakao, K.; Osawa, K.; Ikeno, M.; Yamanaka, S.; Okubo, Y.; Fujimura, K.; Bessho, K. Comparison of human mesenchymal stem cells derived from bone marrow, synovial fluid, adult dental pulp, and exfoliated deciduous tooth pulp. Int. J. Oral Maxillofac. Surg. 2016, 45, 124–131. [Google Scholar] [CrossRef] [PubMed]
  15. 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] [PubMed]
  16. Anoop, M.; Datta, I. Stem Cells Derived from Human Exfoliated Deciduous Teeth (SHED) in Neuronal Disorders: A Review. Curr. Stem Cell Res. Ther. 2021, 16, 535–550. [Google Scholar] [CrossRef]
  17. Relano-Gines, A.; Lehmann, S.; de Periere, D.D.; Hirtz, C. Dental stem cells as a promising source for cell therapies in neurological diseases. Crit. Rev. Cl. Lab. Sci. 2019, 56, 170–181. [Google Scholar] [CrossRef]
  18. Bonaventura, G.; Incontro, S.; Iemmolo, R.; La Cognata, V.; Barbagallo, I.; Costanzo, E.; Barcellona, M.L.; Pellitteri, R.; Cavallaro, S. Dental mesenchymal stem cells and neuro-regeneration: A focus on spinal cord injury. Cell Tissue Res. 2020, 379, 421–428. [Google Scholar] [CrossRef]
  19. Wang, D.; Wang, Y.; Tian, W.; Pan, J. Advances of tooth-derived stem cells in neural diseases treatments and nerve tissue regeneration. Cell Prolif. 2019, 52, e12572. [Google Scholar] [CrossRef] [Green Version]
  20. Xiao, Z.; Lei, T.; Liu, Y.; Yang, Y.; Bi, W.; Du, H. The potential therapy with dental tissue-derived mesenchymal stem cells in Parkinson’s disease. Stem Cell Res. Ther. 2021, 12, 5. [Google Scholar] [CrossRef]
  21. El Moshy, S.; Radwan, I.A.; Rady, D.; Abbass, M.M.S.; El-Rashidy, A.A.; Sadek, K.M.; Dorfer, C.E.; Fawzy El-Sayed, K.M. Dental Stem Cell-Derived Secretome/Conditioned Medium: The Future for Regenerative Therapeutic Applications. Stem Cells Int. 2020, 2020, 7593402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ma, Z.J.; Yang, J.J.; Lu, Y.B.; Liu, Z.Y.; Wang, X.X. Mesenchymal stem cell-derived exosomes: Toward cell-free therapeutic strategies in regenerative medicine. World J. Stem Cells 2020, 12, 814–840. [Google Scholar] [CrossRef]
  23. Khoo, M.L.; Tao, H.; Meedeniya, A.C.; Mackay-Sim, A.; Ma, D.D. Transplantation of neuronal-primed human bone marrow mesenchymal stem cells in hemiparkinsonian rodents. PLoS ONE 2011, 6, e19025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [Green Version]
  26. Kowal, J.; Tkach, M.; Thery, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Tkach, M.; Thery, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
  28. Raposo, G.; Stahl, P.D. Extracellular vesicles: A new communication paradigm? Nat. Rev. Mol. Cell Biol. 2019, 20, 509–510. [Google Scholar] [CrossRef]
  29. Fuloria, S.; Subramaniyan, V.; Dahiya, R.; Dahiya, S.; Sudhakar, K.; Kumari, U.; Sathasivam, K.; Meenakshi, D.U.; Wu, Y.S.; Sekar, M.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Regenerative Potential and Challenges. Biology 2021, 10, 172. [Google Scholar] [CrossRef]
  30. Assoni, A.; Coatti, G.; Valadares, M.C.; Beccari, M.; Gomes, J.; Pelatti, M.; Mitne-Neto, M.; Carvalho, V.M.; Zatz, M. Different Donors Mesenchymal Stromal Cells Secretomes Reveal Heterogeneous Profile of Relevance for Therapeutic Use. Stem Cells Dev. 2017, 26, 206–214. [Google Scholar] [CrossRef]
  31. Yu, S.; Zhao, Y.; Ma, Y.; Ge, L. Profiling the Secretome of Human Stem Cells from Dental Apical Papilla. Stem Cells Dev. 2016, 25, 499–508. [Google Scholar] [CrossRef]
  32. Caseiro, A.R.; Santos Pedrosa, S.; Ivanova, G.; Vieira Branquinho, M.; Almeida, A.; Faria, F.; Amorim, I.; Pereira, T.; Mauricio, A.C. Mesenchymal Stem/Stromal Cells metabolomic and bioactive factors profiles: A comparative analysis on the umbilical cord and dental pulp derived Stem/ Stromal Cells secretome. PLoS ONE 2019, 14, e0221378. [Google Scholar] [CrossRef]
  33. Murakami, M.; Hayashi, Y.; Iohara, K.; Osako, Y.; Hirose, Y.; Nakashima, M. Trophic Effects and Regenerative Potential of Mobilized Mesenchymal Stem Cells from Bone Marrow and Adipose Tissue as Alternative Cell Sources for Pulp/Dentin Regeneration. Cell Transplant. 2015, 24, 1753–1765. [Google Scholar] [CrossRef] [PubMed]
  34. Joo, K.H.; Song, J.S.; Kim, S.; Lee, H.S.; Jeon, M.; Kim, S.O.; Lee, J.H. Cytokine Expression of Stem Cells Originating from the Apical Complex and Coronal Pulp of Immature Teeth. J. Endod. 2018, 44, 87–92.e81. [Google Scholar] [CrossRef] [Green Version]
  35. Nagata, M.; Iwasaki, K.; Akazawa, K.; Komaki, M.; Yokoyama, N.; Izumi, Y.; Morita, I. Conditioned Medium from Periodontal Ligament Stem Cells Enhances Periodontal Regeneration. Tissue Eng. Part A 2017, 23, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kumar, A.; Kumar, V.; Rattan, V.; Jha, V.; Bhattacharyya, S. Secretome proteins regulate comparative osteogenic and adipogenic potential in bone marrow and dental stem cells. Biochimie 2018, 155, 129–139. [Google Scholar] [CrossRef] [PubMed]
  37. Konala, V.B.R.; Bhonde, R.; Pal, R. Secretome studies of mesenchymal stromal cells (MSCs) isolated from three tissue sources reveal subtle differences in potency. In Vitro Cell. Dev. Biol. Anim. 2020, 56, 689–700. [Google Scholar] [CrossRef] [PubMed]
  38. Kim, K.; Jeon, M.; Lee, H.S.; Park, J.C.; Moon, S.J.; Kim, S.O.; Cho, S.W.; Song, J.S. Comparative analysis of secretory factors from permanent- and deciduous-teeth periodontal ligament cells. Arch. Oral Biol. 2016, 71, 65–79. [Google Scholar] [CrossRef] [PubMed]
  39. Ishizaka, R.; Hayashi, Y.; Iohara, K.; Sugiyama, M.; Murakami, M.; Yamamoto, T.; Fukuta, O.; Nakashima, M. Stimulation of angiogenesis, neurogenesis and regeneration by side population cells from dental pulp. Biomaterials 2013, 34, 1888–1897. [Google Scholar] [CrossRef] [PubMed]
  40. Ji, L.; Bao, L.; Gu, Z.; Zhou, Q.; Liang, Y.; Zheng, Y.; Xu, Y.; Zhang, X.; Feng, X. Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells. Immunol. Res. 2019, 67, 432–442. [Google Scholar] [CrossRef]
  41. Silvestro, S.; Chiricosta, L.; Gugliandolo, A.; Pizzicannella, J.; Diomede, F.; Bramanti, P.; Trubiani, O.; Mazzon, E. Extracellular Vesicles Derived from Human Gingival Mesenchymal Stem Cells: A Transcriptomic Analysis. Genes 2020, 11, 118. [Google Scholar] [CrossRef] [Green Version]
  42. Chiricosta, L.; Silvestro, S.; Gugliandolo, A.; Marconi, G.D.; Pizzicannella, J.; Bramanti, P.; Trubiani, O.; Mazzon, E. Extracellular Vesicles of Human Periodontal Ligament Stem Cells Contain MicroRNAs Associated to Proto-Oncogenes: Implications in Cytokinesis. Front. Genet. 2020, 11, 582. [Google Scholar] [CrossRef]
  43. Wang, A.; Liu, J.; Zhuang, X.; Yu, S.; Zhu, S.; Liu, Y.; Chen, X. Identification and Comparison of piRNA Expression Profiles of Exosomes Derived from Human Stem Cells from the Apical Papilla and Bone Marrow Mesenchymal Stem Cells. Stem Cells Dev. 2020, 29, 511–520. [Google Scholar] [CrossRef]
  44. Bousnaki, M.; Bakopoulou, A.; Pich, A.; Papachristou, E.; Kritis, A.; Koidis, P. Mapping the Secretome of Dental Pulp Stem Cells Under Variable Microenvironmental Conditions. Stem Cell Rev. Rep. 2021. [Google Scholar] [CrossRef]
  45. Ahmed, N.E.; Murakami, M.; Kaneko, S.; Nakashima, M. The effects of hypoxia on the stemness properties of human dental pulp stem cells (DPSCs). Sci. Rep. 2016, 6, 35476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Winkel, A.; Jaimes, Y.; Melzer, C.; Dillschneider, P.; Hartwig, H.; Stiesch, M.; von der Ohe, J.; Strauss, S.; Vogt, P.M.; Hamm, A.; et al. Cell culture media notably influence properties of human mesenchymal stroma/stem-like cells from different tissues. Cytotherapy 2020, 22, 653–668. [Google Scholar] [CrossRef]
  47. Bhandi, S.; Al Khatani, A.; Abdulaziz Sumayli, H.; Yahya Sabyei, M.; Mohammed Al Zailai, A.; Ali Sumayli, M.; Ibrahim Hakami, H.; Abdurabu Jafer, M.; Vyas, N.; Ali Baeshen, H.; et al. Comparative analysis of cytokines and growth factors in the conditioned media of stem cells from the pulp of deciduous, young, and old permanent tooth. Saudi J. Biol. Sci. 2021, 28, 3559–3565. [Google Scholar] [CrossRef] [PubMed]
  48. Xia, Y.; Tang, H.N.; Wu, R.X.; Yu, Y.; Gao, L.N.; Chen, F.M. Cell Responses to Conditioned Media Produced by Patient-Matched Stem Cells Derived from Healthy and Inflamed Periodontal Ligament Tissues. J. Periodontol. 2016, 87, e53–e63. [Google Scholar] [CrossRef]
  49. Chin, Y.T.; Liu, C.M.; Chen, T.Y.; Chung, Y.Y.; Lin, C.Y.; Hsiung, C.N.; Jan, Y.S.; Chiu, H.C.; Fu, E.; Lee, S.Y. 2,3,5,4′-tetrahydroxystilbene-2-O-beta-D-glucoside-stimulated dental pulp stem cells-derived conditioned medium enhances cell activity and anti-inflammation. J. Dent. Sci. 2021, 16, 586–598. [Google Scholar] [CrossRef] [PubMed]
  50. Jin, S.; Yang, C.; Huang, J.; Liu, L.; Zhang, Y.; Li, S.; Zhang, L.; Sun, Q.; Yang, P. Conditioned medium derived from FGF-2-modified GMSCs enhances migration and angiogenesis of human umbilical vein endothelial cells. Stem Cell Res. Ther. 2020, 11, 68. [Google Scholar] [CrossRef] [Green Version]
  51. Bhandi, S.; Alkahtani, A.; Mashyakhy, M.; Abumelha, A.S.; Albar, N.H.M.; Renugalakshmi, A.; Alkahtany, M.F.; Robaian, A.; Almeslet, A.S.; Patil, V.R.; et al. Effect of Ascorbic Acid on Differentiation, Secretome and Stemness of Stem Cells from Human Exfoliated Deciduous Tooth (SHEDs). J. Pers. Med. 2021, 11, 589. [Google Scholar] [CrossRef]
  52. Xie, L.; Chen, J.; Ren, X.; Zhang, M.; Thuaksuban, N.; Nuntanaranont, T.; Guan, Z. Alteration of circRNA and lncRNA expression profile in exosomes derived from periodontal ligament stem cells undergoing osteogenic differentiation. Arch. Oral Biol. 2021, 121, 104984. [Google Scholar] [CrossRef]
  53. Liu, T.; Hu, W.; Zou, X.; Xu, J.; He, S.; Chang, L.; Li, X.; Yin, Y.; Tian, M.; Li, Z.; et al. Human Periodontal Ligament Stem Cell-Derived Exosomes Promote Bone Regeneration by Altering MicroRNA Profiles. Stem Cells Int. 2020, 2020, 8852307. [Google Scholar] [CrossRef] [PubMed]
  54. Omi, M.; Hata, M.; Nakamura, N.; Miyabe, M.; Ozawa, S.; Nukada, H.; Tsukamoto, M.; Sango, K.; Himeno, T.; Kamiya, H.; et al. Transplantation of dental pulp stem cells improves long-term diabetic polyneuropathy together with improvement of nerve morphometrical evaluation. Stem Cell Res. Ther. 2017, 8, 279. [Google Scholar] [CrossRef] [Green Version]
  55. Sultan, N.; Amin, L.E.; Zaher, A.R.; Grawish, M.E.; Scheven, B.A. Dental pulp stem cells stimulate neuronal differentiation of PC12 cells. Neural Regen. Res. 2021, 16, 1821–1828. [Google Scholar] [CrossRef] [PubMed]
  56. Gervois, P.; Wolfs, E.; Dillen, Y.; Hilkens, P.; Ratajczak, J.; Driesen, R.B.; Vangansewinkel, T.; Bronckaers, A.; Brone, B.; Struys, T.; et al. Paracrine Maturation and Migration of SH-SY5Y Cells by Dental Pulp Stem Cells. J. Dent. Res. 2017, 96, 654–662. [Google Scholar] [CrossRef] [PubMed]
  57. Ahmed, M.N.; Shi, D.; Dailey, M.T.; Rothermund, K.; Drewry, M.D.; Calabrese, T.C.; Cui, X.T.; Syed-Picard, F.N. Dental Pulp Cell Sheets Enhance Facial Nerve Regeneration via Local Neurotrophic Factor Delivery. Tissue Eng. Part A 2021, 27, 1128–1139. [Google Scholar] [CrossRef]
  58. Chouaib, B.; Collart-Dutilleul, P.Y.; Blanc-Sylvestre, N.; Younes, R.; Gergely, C.; Raoul, C.; Scamps, F.; Cuisinier, F.; Romieu, O. Identification of secreted factors in dental pulp cell-conditioned medium optimized for neuronal growth. Neurochem. Int. 2021, 144, 104961. [Google Scholar] [CrossRef] [PubMed]
  59. Gervois, P.; Ratajczak, J.; Wolfs, E.; Vangansewinkel, T.; Dillen, Y.; Merckx, G.; Bronckaers, A.; Lambrichts, I. Preconditioning of Human Dental Pulp Stem Cells with Leukocyte- and Platelet-Rich Fibrin-Derived Factors Does Not Enhance Their Neuroregenerative Effect. Stem Cells Int. 2019, 2019, 8589149. [Google Scholar] [CrossRef] [Green Version]
  60. Sultan, N.; Amin, L.E.; Zaher, A.R.; Grawish, M.E.; Scheven, B.A. Neurotrophic effects of dental pulp stem cells on trigeminal neuronal cells. Sci. Rep. 2020, 10, 19694. [Google Scholar] [CrossRef]
  61. Nakayama, H.; Iohara, K.; Hayashi, Y.; Okuwa, Y.; Kurita, K.; Nakashima, M. Enhanced regeneration potential of mobilized dental pulp stem cells from immature teeth. Oral Dis. 2017, 23, 620–628. [Google Scholar] [CrossRef] [PubMed]
  62. Yamamoto, T.; Osako, Y.; Ito, M.; Murakami, M.; Hayashi, Y.; Horibe, H.; Iohara, K.; Takeuchi, N.; Okui, N.; Hirata, H.; et al. Trophic Effects of Dental Pulp Stem Cells on Schwann Cells in Peripheral Nerve Regeneration. Cell Transplant. 2016, 25, 183–193. [Google Scholar] [CrossRef]
  63. 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] [PubMed]
  64. 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] [PubMed]
  65. 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]
  66. 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] [PubMed]
  67. Venugopal, C.; Shobha, K.; Rai, K.S.; Pinnelli, V.B.; Kutty, B.M.; Dhanushkodi, A. Neuroprotection by Human Dental Pulp Mesenchymal Stem Cells: From Billions to Nano. Curr. Gene Ther. 2018, 18, 307–323. [Google Scholar] [CrossRef]
  68. Ahmed Nel, M.; Murakami, M.; Hirose, Y.; Nakashima, M. Therapeutic Potential of Dental Pulp Stem Cell Secretome for Alzheimer’s Disease Treatment: An In Vitro Study. Stem Cells Int. 2016, 2016, 8102478. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, J.; Zuzzio, K.; Walker, C.L. Systemic Dental Pulp Stem Cell Secretome Therapy in a Mouse Model of Amyotrophic Lateral Sclerosis. Brain Sci. 2019, 9, 165. [Google Scholar] [CrossRef] [Green Version]
  70. Li, S.; Luo, L.; He, Y.; Li, R.; Xiang, Y.; Xing, Z.; Li, Y.; Albashari, A.A.; Liao, X.; Zhang, K.; et al. Dental pulp stem cell-derived exosomes alleviate cerebral ischaemia-reperfusion injury through suppressing inflammatory response. Cell Prolif. 2021, 54, e13093. [Google Scholar] [CrossRef]
  71. Chen, T.F.; Chen, K.W.; Chien, Y.; Lai, Y.H.; Hsieh, S.T.; Ma, H.Y.; Wang, K.C.; Shiau, C.Y. Dental Pulp Stem Cell-Derived Factors Alleviate Subarachnoid Hemorrhage-Induced Neuroinflammation and Ischemic Neurological Deficits. Int. J. Mol. Sci. 2019, 20, 3747. [Google Scholar] [CrossRef] [Green Version]
  72. Makino, E.; Nakamura, N.; Miyabe, M.; Ito, M.; Kanada, S.; Hata, M.; Saiki, T.; Sango, K.; Kamiya, H.; Nakamura, J.; et al. Conditioned media from dental pulp stem cells improved diabetic polyneuropathy through anti-inflammatory, neuroprotective and angiogenic actions: Cell-free regenerative medicine for diabetic polyneuropathy. J. Diabetes Investig. 2019, 10, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  73. Kanada, S.; Makino, E.; Nakamura, N.; Miyabe, M.; Ito, M.; Hata, M.; Yamauchi, T.; Sawada, N.; Kondo, S.; Saiki, T.; et al. Direct Comparison of Therapeutic Effects on Diabetic Polyneuropathy between Transplantation of Dental Pulp Stem Cells and Administration of Dental Pulp Stem Cell-Secreted Factors. Int. J. Mol. Sci. 2020, 21, 6064. [Google Scholar] [CrossRef]
  74. Jonavice, U.; Tunaitis, V.; Kriauciunaite, K.; Jarmalaviciute, A.; Pivoriunas, A. Extracellular vesicles can act as a potent immunomodulators of human microglial cells. J. Tissue Eng. Regen. Med. 2019, 13, 309–318. [Google Scholar] [CrossRef] [PubMed]
  75. Jonavice, U.; Romenskaja, D.; Kriauciunaite, K.; Jarmalaviciute, A.; Pajarskiene, J.; Kaseta, V.; Tunaitis, V.; Malm, T.; Giniatulin, R.; Pivoriunas, A. Extracellular Vesicles from Human Teeth Stem Cells Trigger ATP Release and Promote Migration of Human Microglia through P2X4 Receptor/MFG-E8-Dependent Mechanisms. Int. J. Mol. Sci. 2021, 22, 970. [Google Scholar] [CrossRef] [PubMed]
  76. 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]
  77. Chen, Y.R.; Lai, P.L.; Chien, Y.; Lee, P.H.; Lai, Y.H.; Ma, H.I.; Shiau, C.Y.; Wang, K.C. Improvement of Impaired Motor Functions by Human Dental Exfoliated Deciduous Teeth Stem Cell-Derived Factors in a Rat Model of Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 3807. [Google Scholar] [CrossRef]
  78. Jarmalaviciute, A.; Tunaitis, V.; Pivoraite, U.; Venalis, A.; Pivoriunas, A. Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine-induced apoptosis. Cytotherapy 2015, 17, 932–939. [Google Scholar] [CrossRef]
  79. Narbute, K.; Pilipenko, V.; Pupure, J.; Dzirkale, Z.; Jonavice, U.; Tunaitis, V.; Kriauciunaite, K.; Jarmalaviciute, A.; Jansone, B.; Klusa, V.; et al. Intranasal Administration of Extracellular Vesicles Derived from Human Teeth Stem Cells Improves Motor Symptoms and Normalizes Tyrosine Hydroxylase Expression in the Substantia Nigra and Striatum of the 6-Hydroxydopamine-Treated Rats. Stem Cells Transl. Med. 2019, 8, 490–499. [Google Scholar] [CrossRef] [Green Version]
  80. 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]
  81. Shimojima, C.; Takeuchi, H.; Jin, S.; Parajuli, B.; Hattori, H.; Suzumura, A.; Hibi, H.; Ueda, M.; Yamamoto, A. Conditioned Medium from the Stem Cells of Human Exfoliated Deciduous Teeth Ameliorates Experimental Autoimmune Encephalomyelitis. J. Immunol. 2016, 196, 4164–4171. [Google Scholar] [CrossRef] [Green Version]
  82. 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. Off. J. Soc. Neurosci. 2015, 35, 2452–2464. [Google Scholar] [CrossRef] [Green Version]
  83. 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] [Green Version]
  84. 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]
  85. Hayashi, Y.; Kato, H.; Nonaka, K.; Nakanishi, H. Stem cells from human exfoliated deciduous teeth attenuate mechanical allodynia in mice through distinct from the siglec-9/MCP-1-mediated tissue-repairing mechanism. Sci. Rep. 2021, 11, 20053. [Google Scholar] [CrossRef] [PubMed]
  86. Kano, F.; Matsubara, K.; Ueda, M.; Hibi, H.; Yamamoto, A. Secreted Ectodomain of Sialic Acid-Binding Ig-Like Lectin-9 and Monocyte Chemoattractant Protein-1 Synergistically Regenerate Transected Rat Peripheral Nerves by Altering Macrophage Polarity. Stem Cells 2017, 35, 641–653. [Google Scholar] [CrossRef]
  87. Sugimura-Wakayama, Y.; Katagiri, W.; Osugi, M.; Kawai, T.; Ogata, K.; Sakaguchi, K.; Hibi, H. Peripheral Nerve Regeneration by Secretomes of Stem Cells from Human Exfoliated Deciduous Teeth. Stem Cells Dev. 2015, 24, 2687–2699. [Google Scholar] [CrossRef] [PubMed]
  88. Li, Y.; Yang, Y.Y.; Ren, J.L.; Xu, F.; Chen, F.M.; Li, A. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats. Stem Cell Res. Ther. 2017, 8, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Inoue, T.; Sugiyama, M.; Hattori, H.; Wakita, H.; Wakabayashi, T.; Ueda, M. Stem cells from human exfoliated deciduous tooth-derived conditioned medium enhance recovery of focal cerebral ischemia in rats. Tissue Eng. Part A 2013, 19, 24–29. [Google Scholar] [CrossRef] [Green Version]
  90. Yamagata, M.; Yamamoto, A.; Kako, E.; Kaneko, N.; Matsubara, K.; Sakai, K.; Sawamoto, K.; Ueda, M. Human dental pulp-derived stem cells protect against hypoxic-ischemic brain injury in neonatal mice. Stroke 2013, 44, 551–554. [Google Scholar] [CrossRef]
  91. Miura-Yura, E.; Tsunekawa, S.; Naruse, K.; Nakamura, N.; Motegi, M.; Nakai-Shimoda, H.; Asano, S.; Kato, M.; Yamada, Y.; Izumoto-Akita, T.; et al. Secreted factors from cultured dental pulp stem cells promoted neurite outgrowth of dorsal root ganglion neurons and ameliorated neural functions in streptozotocin-induced diabetic mice. J. Diabetes Investig. 2020, 11, 28–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tsuruta, T.; Sakai, K.; Watanabe, J.; Katagiri, W.; Hibi, H. Dental pulp-derived stem cell conditioned medium to regenerate peripheral nerves in a novel animal model of dysphagia. PLoS ONE 2018, 13, e0208938. [Google Scholar] [CrossRef] [Green Version]
  93. Rajan, T.S.; Giacoppo, S.; Trubiani, O.; Diomede, F.; Piattelli, A.; Bramanti, P.; Mazzon, E. Conditioned medium of periodontal ligament mesenchymal stem cells exert anti-inflammatory effects in lipopolysaccharide-activated mouse motoneurons. Exp. Cell Res. 2016, 349, 152–161. [Google Scholar] [CrossRef]
  94. Ballerini, P.; Diomede, F.; Petragnani, N.; Cicchitti, S.; Merciaro, I.; Cavalcanti, M.; Trubiani, O. Conditioned medium from relapsing-remitting multiple sclerosis patients reduces the expression and release of inflammatory cytokines induced by LPS-gingivalis in THP-1 and MO3.13 cell lines. Cytokine 2017, 96, 261–272. [Google Scholar] [CrossRef]
  95. Rajan, T.S.; Giacoppo, S.; Diomede, F.; Ballerini, P.; Paolantonio, M.; Marchisio, M.; Piattelli, A.; Bramanti, P.; Mazzon, E.; Trubiani, O. The secretome of periodontal ligament stem cells from MS patients protects against EAE. Sci. Rep. 2016, 6, 38743. [Google Scholar] [CrossRef] [Green Version]
  96. Soundara Rajan, T.; Giacoppo, S.; Diomede, F.; Bramanti, P.; Trubiani, O.; Mazzon, E. Human periodontal ligament stem cells secretome from multiple sclerosis patients suppresses NALP3 inflammasome activation in experimental autoimmune encephalomyelitis. Int. J. Immunopathol. Pharmacol. 2017, 30, 238–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Giacoppo, S.; Thangavelu, S.R.; Diomede, F.; Bramanti, P.; Conti, P.; Trubiani, O.; Mazzon, E. Anti-inflammatory effects of hypoxia-preconditioned human periodontal ligament cell secretome in an experimental model of multiple sclerosis: A key role of IL-37. FASEB J. 2017, 31, 5592–5608. [Google Scholar] [CrossRef] [Green Version]
  98. Rajan, T.S.; Diomede, F.; Bramanti, P.; Trubiani, O.; Mazzon, E. Conditioned medium from human gingival mesenchymal stem cells protects motor-neuron-like NSC-34 cells against scratch-injury-induced cell death. Int. J. Immunopathol. Pharmacol. 2017, 30, 383–394. [Google Scholar] [CrossRef] [PubMed]
  99. Mao, Q.; Nguyen, P.D.; Shanti, R.M.; Shi, S.; Shakoori, P.; Zhang, Q.; Le, A.D. Gingiva-Derived Mesenchymal Stem Cell-Extracellular Vesicles Activate Schwann Cell Repair Phenotype and Promote Nerve Regeneration. Tissue Eng. Part A 2019, 25, 887–900. [Google Scholar] [CrossRef] [Green Version]
  100. Rao, F.; Zhang, D.; Fang, T.; Lu, C.; Wang, B.; Ding, X.; Wei, S.; Zhang, Y.; Pi, W.; Xu, H.; et al. Exosomes from Human Gingiva-Derived Mesenchymal Stem Cells Combined with Biodegradable Chitin Conduits Promote Rat Sciatic Nerve Regeneration. Stem Cells Int. 2019, 2019, 2546367. [Google Scholar] [CrossRef]
  101. Kolar, M.K.; Itte, V.N.; Kingham, P.J.; Novikov, L.N.; Wiberg, M.; Kelk, P. The neurotrophic effects of different human dental mesenchymal stem cells. Sci. Rep. 2017, 7, 12605. [Google Scholar] [CrossRef]
  102. Yu, S.; Zhao, Y.; Fang, T.J.; Ge, L. Effect of the Soluble Factors Released by Dental Apical Papilla-Derived Stem Cells on the Osteo/Odontogenic, Angiogenic, and Neurogenic Differentiation of Dental Pulp Cells. Stem Cells Dev. 2020, 29, 795–805. [Google Scholar] [CrossRef] [PubMed]
  103. Ganz, J.; Arie, I.; Ben-Zur, T.; Dadon-Nachum, M.; Pour, S.; Araidy, S.; Pitaru, S.; Offen, D. Astrocyte-like cells derived from human oral mucosa stem cells provide neuroprotection in vitro and in vivo. Stem Cells Transl. Med. 2014, 3, 375–386. [Google Scholar] [CrossRef] [PubMed]
  104. Gonzalez-Gonzalez, A.; Garcia-Sanchez, D.; Dotta, M.; Rodriguez-Rey, J.C.; Perez-Campo, F.M. Mesenchymal stem cells secretome: The cornerstone of cell-free regenerative medicine. World J. Stem Cells 2020, 12, 1529–1552. [Google Scholar] [CrossRef] [PubMed]
  105. Bogatcheva, N.V.; Coleman, M.E. Conditioned Medium of Mesenchymal Stromal Cells: A New Class of Therapeutics. Bioch. Biokhimiia 2019, 84, 1375–1389. [Google Scholar] [CrossRef]
  106. Pawitan, J.A. Prospect of stem cell conditioned medium in regenerative medicine. BioMed Res. Int. 2014, 2014, 965849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [Green Version]
  108. Chang, C.; Yan, J.; Yao, Z.; Zhang, C.; Li, X.; Mao, H.Q. Effects of Mesenchymal Stem Cell-Derived Paracrine Signals and Their Delivery Strategies. Adv. Healthc. Mater. 2021, 10, e2001689. [Google Scholar] [CrossRef]
  109. Ward, K.R.; Matejtschuk, P. The Principles of Freeze-Drying and Application of Analytical Technologies. Methods Mol. Biol. 2021, 2180, 99–127. [Google Scholar] [CrossRef]
  110. Izutsu, K.I. Applications of Freezing and Freeze-Dry in Pharmaceutical Formulations. Adv. Exp. Med. Biol. 2018, 1081, 371–383. [Google Scholar] [CrossRef] [PubMed]
Table 1. Overview of the main factors found in the dental MSC secretome.
Table 1. Overview of the main factors found in the dental MSC secretome.
MSCsSecretomeContained FactorsComparison with Other MSC
Secretomes
Ref.
SCAPsCM2046 proteins, included chemokines, angiogenic, immunomodulatory, antiapoptotic, and neuroprotective factors, ECM proteins151 proteins were different by at least twofold compared to BMSCs.
SCAPs CM: ↑ proteins involved in metabolic processes, transcription, chemokines and neurotrophins. ↓ proteins linked to biological adhesion, developmental processes, immune function, ECM and pro-angiogenic factors
[31]
DPSCsCMAng-2, EGF, Endoglin, Endothelin-1, Eotaxin-1, FGF-1, FGF-2, Flt-3L, Follistatin, G-CSF, GM-CSF, GRO pan, HB-EGF, HGF, IFNα2, IFNγ, IL-12(p40), IL-12(p70), IL-13, IL-15, IL-1B, IL-5, IL-8, IL-9, IP-10, Leptin, MCP-1, MCP-3, PDGF-AA, PDGF-BB, PLGF, RANTES, TGF-α, TGF-β1, TGF-β2, TGF-β3, TNFα, TNFβ, VEGF-A, VEGF-C, VEGF-DDifferences compared to UC-MSCs at different time points: Eotaxin-1, FGF-2, Fractalkine, GRO pan, IFNα2, IL-1α, IL-6, MCP-1, MCP-3, PDGF-BB, RANTES, TGF-β1, VEGF-A, VEGF-C[32]
DPSCsCMIGF-1, IL10, IGFBP-6, NT-3, BMP-4, MIP-1δ, NAP-2, TGF-β3, TGF-β1, MIP-3α, TNF-α, TNF-β, ICAM-1, NT-4, I-TAC, TARC, Axl, THPO, TECK, Acrp-30, ICAM-3, EGFR, AgRP, XCL-1, MIFUpregulated in DACCs: IGF-1, IL10, IGFBP-6
Downregulated in DACCs: NT-3, BMP-4, MIP-1δ, NAP-2, TGF-β3, TGF-β1, MIP-3α, TNF-α, TNF-β, ICAM-1, NT-4, I-TAC, TARC, Axl, THPO, TECK, Acrp-30, ICAM-3, EGFR, AgRP, XCL-1, MIF
[34]
PDLSCsCM99 proteins, including matrix proteins, enzymes, growth factors, cytokines, and angiogenic factors-[35]
DPSCs, SCAPs and DFSCsCMOsteogenic lineage related proteins were more in Dental MSC secretomeDental MSCs showed more psteogenic protein compared to BMSCs[36]
SHEDsCMFGF-2, IL-10, PDGF, SDF-1, Ang-1, TGF-β3, HGF, INF-γ, VEGF, and IL-6↑ TGF-β3 and angiopoietin-1 in BMSCs, HGF and IFN-γ in SHED, VEGF in WJ-MSC.
PDGF-A, IL-10, FGF-2, and SDF-1 were similar in all MSCs
[37]
Permanent and deciduous-teeth PDLSCsCM76 proteins in permanent-PDLSCs CM, 20 in deciduous-PDLSCs CM, and 19 in both samples;
permanent-PDLSCs CM: proteins involved in cell growth, cell communication, and signal transduction, higher levels of NT-3 and NT-4 and angiogenesis-related cytokines such as EGF and IGF-1.
Deciduous-PDLSCs CM: proteins involved in regulation of the cell cycle, cytokines involved in immune response and in tissue degradation and catalytic activities, including MMP1, PSMA1, and CUL7
-[38]
GMSCsEVsTranscripts for growth factors such as TGF-β, FGF, VEGF, GDNF family ligands and neurotrophins, such as NGF, BDNF, NT-3 and NT-4, ILs and members of the Wnt family-[41]
PDLSCsEVsContained non-coding RNA: antisense RNA, long non-coding RNA, miRNAs (MIR24-2, MIR142, MIR335, MIR490, and MIR296)-[42]
SCAPsEXOs593 piRNAs920 piRNAs in BMSC-EXOs. 21
piRNAs were differentially expressed
[43]
DPSCsCMCM obtained in normoxic conditions: ↑ molecules with anti-inflammatory, tissue repair and regenerative properties compared to hypoxic CM-[44]
SHEDs, young DPSCs, old DPSCsCMIL-4, IL-2, CXCL10, IL-1B, TNF-A, CCL2, IL-17A, IL-6, IL-10, IFN-γ, IL-12P17, CXCL8, TGF-β1, ANG-2, EGF, EPO, BFGF, G-CSF, GM-CSF, HGF, M-CSF, PDGF-AA, PDGF-BB, SCF, TGF-α, VEGF
SHEDs and young DPSCs: ↑ growth factors, ↓ pro-inflammatory cytokines
-[47]
DPSCs treated with THSGCMTreatment increased: AKT2, persephin, NGFR, PTHrP, maspin, leptin, STAT3, YES1, MMP-13, FGF-5, HER3, FGF-16, IGF-BP1, LH, myostatin, HDAC1, SDF-1β, MDC, MCP-4, L-selectin, TNF-α, STAT6, β-2-MICROGLOBULIN, APRIL, eotaxin-3, MCP-1, LIGHT, galectin 3, LD78β, MIP-1β, granzyme B, LEC.
Treatment reduced: TSH, EGF, CTGFL, HGH, FSH, vaspin, PDGF-AA, GM-CSF, KGF, FGF-acidic, FAK, GDF-3, TGF-β2, IGF-I, MMP-2, STAT-1, TIMP-1, eotaxin, MMP-23, IL-12, galectin-1, ena-78, IL-15, IL-4, IL-22, IL-6, IL-13, IL-1β
-[49]
FGF-2-modified GMSCsCM↑ VEGF-A, FGF-2, TGF-β.-[50]
SHEDs treated with Ascorbic acidCMTreatment ↑ VEGF, TGF-α, SCF, TGF-β, IGF-1, HGF, bFGF, Ang-1, EGF, Ang-2, TNF-α, IL-10, IL-6, IL-17A, NO, IDO, SDF-1, PGE-2.
Treatment ↓ CCL2, TGF-β1
-[51]
Undifferentiated PDLSCs or PDLSCs exposed to osteogenic differentiation mediumEVs69–557 circRNAs and 2907–11,581 lncRNAs. After 5 and 7 days of exposure to differentiation medium: ↑ 3 circRNAs and 2 lncRNAs, ↓ 39 circRNAs and 5 lncRNAs-[52]
Undifferentiated PDLSCs or osteogenic differentiated PDLSCsEXOS72 upregulated miRNAs, 35 downregulated miRNAs after osteogenic induction-[53]
Ang, angiopoietin; BDNF, brain-derived neurotrophic factor; BMP, bone morphogenetic protein; BMSCs, bone marrow MSCs; circRNA, circular RNA; CM, conditioned medium; CUL7, cullin 7; CXCL, C-X-C motif chemokine ligand; DACCs, developing apical complex cells; DFSCs, dental follicle stem cells; DPSCs, dental pulp stem cells; ECM, extracellular matrix; EGF, epidermal growth factor; ECM, extracellular matrix; EVs, extracellular vesicles; EXOs, exosomes; FGF, fibroblast growth factor; G-CSF, granulocyte colony-stimulating factor; GDNF, glial-cell-derived neurotrophic factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; GMSCs, gingival MSCs; HGF, hepatocyte growth factor; ICAM, Intercellular Adhesion Molecule; IDO, indoleamine 2,3-dioxygenase; IFN, interferon; IGF, insulin-like growth factor; IL, interleuchin; lncRNA, long noncoding RNA; MCP, Monocyte Chemoattractant Protein; miRNA, microRNA; MMP, matrix metalloproteinase; NGF, nerve growth factor; NT, neurotrophin; PDGF, platelet-derived growth factor; PDLSCs, periodontal ligament stem cells; piRNA, PIWI-interacting RNAs; PSMA1, Proteasome subunit, alpha type; SCAPs, stem cells from apical papilla; SDF, stromal cell–derived factor; SHEDs, stem cells from human exfoliated deciduous teeth; TGF, transforming growth factor; THSG, 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside; TIMP, tissue inhibitor of metalloproteinase; TNF, Tumor Necrosis Factor; UC-MSCs, umbilical cord mesenchymal stem cells; VEGF, vascular endothelial growth factor ↑, increase/improvements; ↓, reduction.
Table 2. Overview of studies involving dental pulp stem cell secretome.
Table 2. Overview of studies involving dental pulp stem cell secretome.
Secretome TypeAdministrationPreclinical ModelFactors Contained in SecretomeResultsRef.
CM-Dorsal root ganglion neurons; Schwann cell-↑ neurite outgrowth, Schwann cell viability and myelin formation[54]
CM-PC12 cellsNGF, BDNF, GDNF and NT-3↑ survival and neurite outgrowth[55]
CM-SH-SY5Y cell line-↑ neurite outgrowth, neuronal markers and voltage-gated Ca2+ channels[56]
CM obtained by DPSC sheet-Neuronally differentiated SH-SY5Y neuroblastoma cellsBDNF, GDNF, NT-3↑ formation and outgrowth of neurites[57]
CM with and without B-27 supplement-Primary sensory neuronsOnly with B-27: GDF-15, SCF R, Insulin;
Only without B-27: FGF-4, GH, and VEGF-D;
In common: NT-3, PDGF-AA, HGF, IGFBP (1–6), EGFR, OPG, VEGF, BMP-7, FGF-7, and IGF-1.
↑ neurite outgrowth, B-27 supplement enhanced the effect[58]
CM-Neural stem cells-↑ neuritogenesis[59]
CM-Primary trigeminal ganglion neuronal cellsNGF, BDNF, NT-3 and GDNF↑ survival, extensive neurite outgrowth and branching.[60]
CM obtained by basal and G-CSF-mobilized DPSCs-TGW human neuroblastomacells-CM from mobilized DPSCs ↑ neurite extension[61]
CM obtained by G-CSF-mobilized DPSCs-Neuronal Schwann RT4-D6P2T cells-↑ proliferation and migratory activity[62]
CM from SHEDs and DPSCs-Cerebral granular neurons with axon growth inhibitors-↑ regeneration inhibiting axon growth inhibitors[63]
CM from DPSCs, SCAPs and DFSCs-Preneuroblastic cell line IMR-32 cellsGCSF, IFN-γ, TGF-β, NGF, BDNF and NT-3↑ neural differentiation.
Neurite length was higher with DPSC CM.
[64]
DPSC, BMSC and AMSC coculture-Retinal ganglion cellsNGF, BDNF, NT-3, VEGF, GDNF, PDGF-AA↑ survival.
DPSCs released higher levels of NGF, BDNF and VEGF
[65]
CM-Astrocytes exposed to OGD-↑ protective effects.
↓ GFAP, nestin, and musashi-1 expression, ROS and IL-1β
[66]
EXOs, CM or neuron–MSC-co-culture from DPSCs or BMSCs-Hippocampal cell line (H3) exposed to kainic acid-↑ neuroprotection. CM fraction in the range 3–10 kDa showed neuroprotection[67]
CM-Human neuroblastoma SH-SY5Y cells treated with A𝛽1–42VEGF, RANTES, FRACTALKINE, FLT-3, GM-CSF, MCP-1, neprilysin↓A𝛽 cytotoxicity and apoptosis.
↑ cell viability.
DPSC secretome proteolytically degrade A𝛽1–42.
[68]
CMIntraperitoneally; early pre-symptomatic stage: at postnatal day 35–47; late pre-symptomatic stages: postnatal day 70–91; at symptom onset through to end-stageTransgenic mice B6SJL-Tg (SOD1G93A)1 Gur/J-↑ neuromuscular junction innervation; neuromuscular junction preservation, and motor neuron survival, lifespan[69]
EXOsIntravenous singular injection after reperfusionC57BL/6 mice subjected to tMCAO injury followed by reperfusion.OGD/R induced BV2 cells-In vivo: ↓ brain oedema, cerebral infarction, neurological impairment and pro-inflammatory cytokines.
In vitro: ↓ inflammation
[70]
CMIntrathecal injection 10 min before aSAHAneurysmal subarachnoid hemorrhage induced in Wistar ratsIGF-1, TGF-β, TIMP1, TIMP2↑ oxygenation of injured brain, cognitive and motor function.
↓ neuroinflammation
[71]
CMHindlimb skeletal muscles 8 weeks after streptozotocin injectionSprague Dawley rats treated with Streptozotocin-↑ sciatic motor/sensory nerve conduction velocity, sciatic nerve blood flow and intraepidermal nerve fiber density[72]
CMIn the unilateral hindlimb skeletal musclesSprague Dawley rats treated with StreptozotocinVEGF-C, BDNF, IL-1 β,
IL-4, TLR4 and others
↑ sciatic nerve conduction velocity, sciatic nerve blood flow.[73]
Aβ, amyloid β; aSAH, aneurysmal subarachnoid hemorrhage; AMSCs, adipose tissue derived MSCs; BDNF, brain-derived neurotrophic factor; BMP, bone morphogenetic protein; BMSCs, bone marrow MSCs; CM, conditioned medium; DFSCs, dental follicle stem cells; DPSCs, dental pulp stem cells; EXOs, exosomes; FGF, fibroblast growth factor; GDNF, glial-cell-derived neurotrophic factor; GFAP, glial fibrillary acidic protein; G-CSF, granulocyte colony-stimulating factor; HGF, hepatocyte growth factor; IFN, interferon; IGF, insulin-like growth factor; IGFBP, insulin-like growth factor binding protein; IL, interleukin; MCP, Monocyte Chemoattractant Protein; MSCs, mesenchymal stem cells; NGF, nerve growth factor; NT, neurotrophin; OGD/R, oxygen-glucose deprivation–reperfusion; OPG, osteoprotegerin; PDGF, platelet-derived growth factor; ROS, reactive oxygen species; SCAPs, stem cells from apical papilla; SHEDs, stem cells from human exfoliated deciduous teeth; TGF, transforming growth factor; TIMP, tissue inhibitor of metalloproteinase; TLR, Toll like receptor; tMCAO, transient middle cerebral artery occlusion; VEGF, vascular endothelial growth factor; ↑, increase/improvements; ↓, reduction.
Table 3. Overview of studies involving SHED secretome.
Table 3. Overview of studies involving SHED secretome.
Secretome TypeAdministrationPreclinical ModelFactors Contained in SecretomeResultsRef.
EVs-SV40 human microglial cell line treated with LPS or polarized toward M1 or M2-↓ NF-κB signaling
↑ glycolytic activity in M0, M1, and M2 cells
[74]
EVs-Human microglial cells-↑ intracellular Ca2+ and ATP release, motility through P2X4 receptor/milk fat globule-epidermal growth factor-factor VIII (MFG-E8)-dependent mechanisms[75]
CM from DAergic-neuron-like differentiated and undifferentiated SHEDs-Cerebellar granule neurons treated with 6-OHDA-CM obtained from differentiated SHEDs protect primary neurons against 6-OHDA toxicity and accelerated neurite outgrowth[76]
CMIntravenous injectionLewis rats treated with rotenoneIGFBP-6, TIMP-2, TIMP-1, TGF-β1, IGFBP-2, IGFBP-4, BMP-5100 µg/mL of SHED-CM induced the maximal improvement of motor deficits
↑ tyrosine hydroxylase
↓ synuclein levels, Iba-1 positive cells and CD4 levels
[77]
EXOs and MVs from SHEDs grown in standard culture conditions or on laminin-coated three-dimensional alginate micro-carriers-Dopaminergic neurons differentiated from ReNcell VM human neural stem cells treated with 6-OHDA-EXOs, but not MVs, derived from SHEDs grown on laminin-coated three-dimensional alginate micro-carriers suppressed apoptosis[78]
EVsIntranasalMale Wistar rats treated with 6-OHDA-↑ motor function[79]
CMIntranasally 24 h after i.c.v. injection of A𝛽1−40Mice receiving an i.c.v. injection of A𝛽1−40 peptide-↑ cognitive function and neurotrophic factors. ↓ oxidative stress[80]
CMIntravenous at day 14 postimmunization (peak of EAE)EAE induced C57BL/6J miceED–Siglec-9 and HGF↓ demyelination and axonal injury, inflammatory cell infiltration and proinflammatory cytokine expression[81]
CMIntrathecallySprague Dawley rats with spinal cord contusion injury79 proteins of which 28 involved in neuroregenerative processes; MCP-1 and ED-Siglec-9 may be involved in M2-like macrophage differentiation↑ functional recovery
↓ pro-inflammatory cytokines
[82]
CM alone or loaded on collagen hydrogelIntraspinal injectionSprague Dawley rats subjected to SCI-CM on collagen hydrogel ↑ functional recovery[83]
CM alone or loaded on collagen hydrogelIntraspinal injectionSprague Dawley rats subjected to SCI-CM on collagen hydrogel: ↑ volume of preserved white and gray matter and the total number of neurons and oligodendrocytes
↓ lesion volume and lesion length
[84]
CMIntravenous administration 5 days after peripheral nerve injuryMale C57BL/6J mice subjected to peripheral nerve injury made by the transection of the L4 spinal nerve-↓ allodynia and activation of microglia and astrocytes[85]
CMCM loaded on the collagen sponge placed in the nerve gapFemale Sprague Dawley with rat facial nerves transectionMCP-1 and ED-Siglec-9↑ neurological function[86]
CMSilicon conduits containing CMSchwann cells and dorsal root ganglia; male Wistar/ST rats subjects to sciatic nerve transectionNGF, BDNF, NT-3, GDNF, CNTF, VEGF, and HGFIn vitro: ↑ proliferation, migration, and expression of neuron-, ECM-, and angiogenesis-related genes, neurite outgrowth of dorsal root ganglia and increased cell viability
In vivo: ↑ axon regeneration, myelination, motor function.
↓ muscle atrophy
[87]
EXOs-BV-2 cells treated with LPS; Wistar rats subjected to TBI-↑ motor function[88]
CMIntranasally 3 days after MCAOSprague Dawley rats subjected to permanent MCAO-↓ motor disability and infarct volume[89]
CMIntracerebral administration 24 h after hypoxia-ischemiaICR mice with hypoxia-ischemia brain injury-↑ neurological function and survival rate[90]
CMInjection into soleus muscles twice a week over a period of 4 weeks twelve weeks after the induction of diabetesDorsal root ganglion neurons; C57BL/6 mice treated with streptozotocinNGF, BDNF, FGF2 and VEGF in fraction > 6 kDaIn vitro: ↑ neurite outgrowth
In vivo: ↓ decline in sensory nerve conduction velocities
[91]
CMIntravenous simultaneously with the superior laryngeal nerve damageMale Wistar/ST rats subjected to superior laryngeal nerve damage-↑ functional recovery, myelination and axonal regeneration[92]
6-OHDA, 6-hydroxy-dopamine; Aβ, amyloid β; BDNF, brain-derived neurotrophic factor; BMP, bone morphogenetic protein; CM, conditioned medium; CNTF, ciliary neurotrophic factor; EAE, experimental autoimmune encephalomyelitis; ECM, extracellular matrix; ED-Siglec-9, ectodomain of sialic-acid-binding Ig-like lectin-9; EXOs, exosomes; EVs, extracellular vesicles; FGF, fibroblast growth factor; GDNF, glial-cell-derived neurotrophic factor; HGF, hepatocyte growth factor; i.c.v., intracerebroventricular; IGFBP, insulin-like growth factor binding protein; LPS, lipopolysaccharide; MCP, Monocyte Chemoattractant Protein; MVs, microvesicles; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; NGF, nerve growth factor; NT, neurotrophin; OGD, oxygen-glucose deprivation; SHEDs, stem cells from human exfoliated deciduous teeth; MCAO, middle cerebral artery occlusion; SCI, spinal cord injury; TBI, traumatic brain injury; TGF, transforming growth factor; TIMP, tissue inhibitor of metalloproteinase; VEGF, vascular endothelial growth factor; ↑, increase/improvements; ↓, reduction.
Table 4. Overview of studies involving PDLSC secretome.
Table 4. Overview of studies involving PDLSC secretome.
Secretome TypeAdministrationPreclinical ModelFactors Contained in
Secretome
ResultsRef.
CM obtained from PDLSCs of RR-MS patients-NSC-34 neurons treated with the medium of Lipopolysaccharide-stimulated RAW macrophageIL-10, TGF-β↓ inflammation and apoptosis
↑ IL-10 and neuroprotective markers
[93]
CM from PDLSCs of healthy subjects or RR-MS patients-PMA differentiated THP-1 cells, undifferentiated and PMA-differentiated MO3.13 cells treated with Porphyromonas gingivalis LPS-↓ TNFα, IL-1β and IL-6 levels[94]
CM and EXOs/MVs (EMVs) from PDLSCs obtained from RR-MS patients and healthy donorsIntravenous at disease onsetEAE induced C57BL/6 miceIL-10 and TGF-β↑ remyelination of spinal cord
↓ inflammation in spinal cord and spleen and apoptosis
[95]
CM and EXOs/MVs (EMVs) from PDLSCs obtained from RR-MS patientsIntravenous at disease onsetEAE induced C57BL/6 miceIL-10, SDF-1α, TGF-β, IL-15, MCP-1 and MIP-1α↓ NALP3 inflammasome activation, TLR-4 and NF-κB[96]
CM obtained from PDLSCs cultured in hypoxic conditionsIntravenous 14 days after EAE inductionEAE induced C57BL/6 mice; Scratch injured murine motor-neuron-like NSC-34 cellsNT3, IL-10 and TGF-βIn vivo: ↓ clinical and histologic disease score, inflammatory cell infiltration, oxidative stress and apoptosis↑ remyelination in spinal cord, IL-37, activation of the PI3K/Akt/mTOR pathway.
In vitro: ↓ inflammation, oxidative stress, and apoptosis
[97]
CM, conditioned medium; EAE, experimental autoimmune encephalomyelitis; EMVs, EXOs/MVs; EXOs, exosomes; IL, interleukin; MCP, Monocyte Chemoattractant Protein; MIP, Macrophage Inflammatory Protein; MVs, microvesicles; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; NT, neurotrophin; PMA, Phorbol 12-myristate 13-acetate; PDLSCs, periodontal ligament stem cells; RR-MS, relapsing remitting multiple sclerosis; SDF-1α, stromal cell–derived factor 1α; TGF, transforming growth factor; TLR, Toll like receptor; TNF, Tumor Necrosis Factor; ↑, increase/improvements; ↓, reduction.
Table 5. Overview of studies involving other dental MSCs secretome.
Table 5. Overview of studies involving other dental MSCs secretome.
MSCsSecretome TypeAdministrationPreclinical ModelFactors Contained in
Secretome
ResultsRef.
GMSCsCM-Scratch injured murine motor-neuron-like NSC-34 cellsNGF, NT-3, IL-10 and TGF-β↓ apoptosis, oxidative stress, TNF-α;
↑ IL-10, BDNF and NT-3
[98]
GMSCsEVsGelfoam mixed with EVs wrapped around the
injury site
C57BL/6J mice subjected to crush injury of sciatic nerve; rat Schwann cell line RT4-D6P2T-In vivo: ↑ functional recovery and axonal regeneration, proliferation and migration of Schwann cells.
In vitro: ↑ expression of Schwann cell dedifferentiation/repair phenotype-related genes
[99]
GMSCsEXOsChitin conduit enriched with EXOsDRG cell co-cultured with Schwann cell; SD rats subjected to sciatic nerve defect-In vitro: ↑ Schwann cell proliferation, DRG axon growth.
In vivo: ↑ number and diameter of nerve fibers, myelin formation, nerve conduction, muscle function and motor function
[100]
SCAPs, DPSCs and PDLSCsCM-Retinoic acid differentiated SH-SY5Y cellsBDNF, NGF, NT-3, GDNF, VEGF-A and angiopoietin-1↑ the percentage of cells producing neurites and the total neurite outgrowth length[101]
SCAPsCM-DPSCs cultured in medium for neural stem cell growth-↑ neurogenic effect compared to BMSCs-CM, neurogenic markers[102]
Astrocyte-like differentiated OMSCsCM-Mouse motor neuron-like NSC-34 cells exposed to hypoxia or hydrogen peroxide-↑ cell viability[103]
BDNF, brain-derived neurotrophic factor; BMSCs, bone marrow MSCs; CM, conditioned medium; DPSCs, dental pulp stem cells; DRG, dorsal root ganglion; EVs, extracellular vesicles; EXOs, exosomes; GDNF, glial-cell-derived neurotrophic factor; GMSCs, gingival MSCs; IL, interleukin; NGF, nerve growth factor; NT, Neurotrophin; OMSCs, oral mucosa stem cells; PDLSCs, periodontal ligament stem cells; SCAPs, stem cells from apical papilla; TGF, transforming growth factor; TNF, Tumor Necrosis Factor; VEGF, vascular endothelial growth factor; ↑, increase/improvements; ↓, reduction.
Table 6. Studies analyzing proteome, transcriptome, and non-coding RNA profiles of different dental MSCs.
Table 6. Studies analyzing proteome, transcriptome, and non-coding RNA profiles of different dental MSCs.
MSCs TypeSecretomeProfileRef.
SCAPsCMProteome[31]
DPSCsCMMetabolomic and bioactive factors profiles[32]
PDLSCsCMProteome[35]
DPSCs, SCAPs, DFSCsCMProteome[36]
PDLSCsCMProteome[38]
GMSCsEVsTranscriptome[41]
PDLSCsEVsNon-coding RNA[42]
SCAPsEXOspiRNA profile[43]
PDLSCsEVscircRNA and lncRNA profile[52]
PDLSCsEXOsmiRNA profile[53]
circRNAs, circular RNA; CM, conditioned medium; DFSCs, dental follicle stem cells; DPSCs, dental pulp stem cells; EVs, extracellular vesicles; EXOs, exosomes; GMSCs, gingival MSCs; lncRNAs, long non-coding RNAs; miRNA, microRNA; PDLSCs, periodontal ligament stem cells; piRNA, PIWI-interacting RNAs; SCAPs, stem cells from apical papilla.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gugliandolo, A.; Mazzon, E. Dental Mesenchymal Stem Cell Secretome: An Intriguing Approach for Neuroprotection and Neuroregeneration. Int. J. Mol. Sci. 2022, 23, 456. https://doi.org/10.3390/ijms23010456

AMA Style

Gugliandolo A, Mazzon E. Dental Mesenchymal Stem Cell Secretome: An Intriguing Approach for Neuroprotection and Neuroregeneration. International Journal of Molecular Sciences. 2022; 23(1):456. https://doi.org/10.3390/ijms23010456

Chicago/Turabian Style

Gugliandolo, Agnese, and Emanuela Mazzon. 2022. "Dental Mesenchymal Stem Cell Secretome: An Intriguing Approach for Neuroprotection and Neuroregeneration" International Journal of Molecular Sciences 23, no. 1: 456. https://doi.org/10.3390/ijms23010456

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