Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(1): e23738, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38226277

ABSTRACT

Adverse environmental stress causes depressive symptoms with the impairments of memory formation, cognition, and motivation, however, their underlying neural bases have not been well understood, especially based on the observation of living animals. In the present study, therefore, the mice model of restraint-induced stress was examined electrophysiologically to investigate the alterations of hippocampal sharp wave ripples (SWRs) and theta rhythms. In addition, the therapeutic effects of physical exercise on the amelioration of those hippocampal impairments were examined in combination with a series of behavioral tests. The data demonstrated that chronic restraint stress caused the reductions of occurrence and amplitude of hippocampal SWRs and the decreases of occurrence, duration, and power of theta rhythms, while physical exercise significantly reverted them to the levels of healthy control. Furthermore, hippocampal adult neurogenesis and microglial activation, previously reported to be involved in the etiology of depression, were histologically examined in the mice. The results showed that the impairment of neurogenesis and alleviation of microglial activation were induced in the depressed mice. On the other hand, physical exercise considerably ameliorated those pathological conditions in the affected brain. Consistently, the data of behavioral tests in mice suggested that physical exercise ameliorated the symptomatic defects of motivation, memory formation, and cognition in the depressed mice. The impairments of hippocampal SWRs and theta rhythms in the affected hippocampus are linked with the symptomatic impairments of cognition and motivation, and the defect of memory formation, respectively, in depression. Taken together, this study demonstrated the implications of impairment of the hippocampal SWRs and theta rhythms in the etiology of depression and their usefulness as diagnostic markers of depression.

2.
Stem Cells Dev ; 29(2): 63-74, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31801412

ABSTRACT

Recently, cell therapy has been developed as a novel treatment for perinatal hypoxic-ischemic encephalopathy (HIE), which is an important cause of neurological disorder and death, and stem cells from human exfoliated deciduous teeth (SHED) express early markers for mesenchymal and neuroectodermal stem cells. We investigated the treatment effect of SHED for HIE in neonatal rats. Seven-day-old rats underwent ligation of the left carotid artery and were exposed to 8% hypoxic treatment. SHED (1 × 105 cells) were injected via the right external jugular vein 24 h after the insult. The effect of intravenous administration of SHED cells was evaluated neurologically and pathophysiologically. In the evaluation of engraftment using quantum dots 655, only a few SHED were detected in the injured cortex. In the immunohistological evaluation 24 h after injection, the numbers of positive cells of active caspase-3 and anti-4 hydroxynonenal antiserum were lower in the SHED group than in the vehicle group. The number of Iba-1+ cells in the cortex was higher in the SHED group. However, the proportion of M1 microglia (Iba-1+/ED-1+) was significantly decreased, whereas M2 microglia (Iba-1+/CD206+) tended to increase in the SHED group. In the behavioral tests performed 5 months after hypoxic treatment, compared to the vehicle group, the SHED group showed significant elongation of the endurance time in the rotarod treadmill test, significantly ameliorated proportion of using the impaired hand in the cylinder test, significantly lower ratio of right/left front paw area in gait analysis, and significantly higher avoidance rate in the active avoidance test. In the in vitro experiment with cultured neurons exposed to oxygen-glucose deprivation, we confirmed the neuroprotective effect of the condition medium of SHED. These results suggested that intravenous administration of SHED exerted a treatment effect both histologically and functionally, possibly via a paracrine effect.


Subject(s)
Hypoxia-Ischemia, Brain/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Tooth, Deciduous/cytology , Administration, Intravenous , Animals , Animals, Newborn , Avoidance Learning/physiology , Cells, Cultured , Child , Disease Models, Animal , Humans , Hypoxia-Ischemia, Brain/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Microglia/metabolism , Motor Activity/physiology , Rats, Wistar , Transplantation, Heterologous/methods , Treatment Outcome
3.
J Tissue Eng Regen Med ; 11(6): 1888-1896, 2017 06.
Article in English | MEDLINE | ID: mdl-28586545

ABSTRACT

In acute liver failure (ALF), a poorly controlled innate immune response causes massive hepatic destruction, which elicits a systemic inflammatory response, progressive multiple organ failure and ultimate sudden death. Although the liver has inherent tissue-repairing activities, its regeneration during ALF fails, and orthotopic liver transplantation is the only curative approach. Here we show that a single intravenous administration of stem cells derived from human exfoliated deciduous teeth (SHEDs) or of SHED-derived serum-free conditioned medium (SHED-CM) into the d-galactosamine-induced rat model of ALF markedly improved the condition of the injured liver and the animals' survival rate. The engraftment of infused SHEDs was very low, and both SHEDs and SHED-CM exerted similar levels of therapeutic effect, suggesting that the SHEDs reversed ALF by paracrine mechanisms. Importantly, SHED-CM attenuated the ALF-induced pro-inflammatory response and generated an anti-inflammatory/tissue-regenerating environment, which was accompanied by the induction of anti-inflammatory M2-like hepatic macrophages. Secretome analysis by cytokine antibody array revealed that the SHED-CM contained multiple tissue-regenerating factors with known roles in anti-apoptosis/hepatocyte protection, angiogenesis, macrophage differentiation and the proliferation/differentiation of liver progenitor cells. Taken together, our findings suggest that SHEDs produce factors that provide multifaceted therapeutic benefits for AFL. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Liver Failure, Acute , Liver Regeneration , Stem Cell Transplantation , Stem Cells/metabolism , Tooth, Deciduous , Animals , Apoptosis , Cell Differentiation , Female , Hepatocytes/metabolism , Hepatocytes/pathology , Heterografts , Humans , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Liver Failure, Acute/therapy , Macrophages/metabolism , Macrophages/pathology , Neovascularization, Physiologic , Rats , Rats, Sprague-Dawley , Stem Cells/pathology
4.
Sci Rep ; 7: 44043, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272428

ABSTRACT

Effective treatments for acute liver failure (ALF) are still lacking. We recently reported that a single intravenous administration of serum-free conditioned medium from stem cells derived from human exfoliated deciduous teeth (SHED-CM) into the D-galactosamine (D-Gal)-induced rat ALF model improves the liver injury. However, the specific factors in SHED-CM that are responsible for resolving ALF remain unclear. Here we found that depleting SHED-CM of two anti-inflammatory M2 macrophage inducers-monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9)-abolished its ability to resolve rat ALF. Furthermore, treatment with MCP-1/sSiglec-9 alone dramatically improved the survival of ALF rats. This treatment induced anti-inflammatory M2, suppressed hepatocyte apoptosis, and promoted hepatocyte proliferation. Treatment with an M2-depletion reagent (mannosylated clodronate liposomes) suppressed the recovery. In addition, MCP-1 and sSiglec-9 synergistically promoted the M2 differentiation of bone marrow-derived macrophages via CCR2, accompanied by the production of multiple liver-regenerating factors. The conditioned medium from MCP-1/sSiglec-9-activated M2 macrophages, but not from interleukin-4-induced ones, suppressed the D-Gal- and LPS-induced apoptosis of primary hepatocytes and promoted their proliferation in vitro. The unique combination of MCP-1/sSiglec-9 ameliorates rat ALF by inhibiting hepatocellular apoptosis and promoting liver regeneration through the induction of anti-inflammatory/tissue-repairing M2 macrophages.


Subject(s)
Chemokine CCL2/metabolism , Liver Failure, Acute/metabolism , Macrophages/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Apoptosis , Cell Proliferation , Culture Media, Serum-Free , Female , Inflammation/complications , Inflammation/metabolism , Liver Failure, Acute/complications , Rats, Sprague-Dawley , Stem Cells/physiology , Tooth Exfoliation , Tooth, Deciduous/cytology
5.
Stem Cells ; 35(3): 641-653, 2017 03.
Article in English | MEDLINE | ID: mdl-27862629

ABSTRACT

Peripheral nerves (PNs) exhibit remarkable self-repairing reparative activity after a simple crush or cut injury. However, the neuronal transection involving a nerve gap overwhelms their repairing activity and causes persistent paralysis. Here, we show that an implantation of the serum-free conditioned medium from stem cells from human exfoliated deciduous teeth (SHED-CM) immersed in a collagen sponge into the nerve gap formed by rat facial nerves transection restored the neurological function. In contrast, SHED-CM specifically depleted of a set of anti-inflammatory M2 macrophage inducers, monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9) lost the ability to restore neurological function in this model. Notably, the combination of MCP-1 and sSiglec-9 induced the polarization of M2 macrophages in vitro, resulting in the expression of multiple trophic factors that enhanced proliferation, migration, and differentiation of Schwann cells, blood vessel formation, and nerve fiber extension. Furthermore, the implantation of a collagen graft containing MCP-1/sSiglec-9 into the nerve gap induced anti-inflammatory M2 macrophage polarization, generated a Schwann-cell bridge instead of fibrotic scar, induced axonal regrowth, and restored nerve function. The specific elimination of M2 macrophages by Mannosylated-Clodrosome suppressed the MCP-1/sSiglec-9-mediated neurological recovery. Taken together, our data suggest that MCP-1/sSiglec-9 regenerates PNs by inducing tissue-repairing M2 macrophages and may provide therapeutic benefits for severe peripheral nerve injuries. Stem Cells 2017;35:641-653.


Subject(s)
Cell Polarity , Chemokine CCL2/metabolism , Macrophages/pathology , Peripheral Nerves/pathology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Child , Facial Nerve/physiopathology , Female , Ganglia, Spinal/metabolism , Humans , Inflammation/pathology , Macrophages/metabolism , Nerve Regeneration , Neuronal Outgrowth , Peripheral Nerves/physiopathology , Rats, Sprague-Dawley , Recovery of Function , Schwann Cells , Signal Transduction , Stem Cells/metabolism , Tooth, Deciduous/cytology
6.
Cytotherapy ; 17(8): 1119-29, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031744

ABSTRACT

BACKGROUND AIMS: Acute respiratory distress syndrome (ARDS) is a severe inflammatory disorder characterized by acute respiratory failure, resulting from severe, destructive lung inflammation and irreversible lung fibrosis. We evaluated the use of stem cells derived from human exfoliated deciduous teeth (SHEDs) or SHED-derived serum-free conditioned medium (SHED-CM) as treatments for bleomycin (BLM)-induced mice acute lung injury (ALI), exhibiting several pathogenic features associated with the human disease ARDS. METHODS: Mice with BLM-induced ALI with or without SHED or SHED-CM treatment were examined for weight loss and survival. The lung tissue was characterized by histological and real-time quantitative polymerase chain reaction analysis. The effects of SHED-CM on macrophage differentiation in vitro were also assessed. RESULTS: A single intravenous administration of either SHEDs or SHED-CM attenuated the lung injury and weight loss in BLM-treated mice and improved their survival rate. Similar recovery levels were seen in the SHEDs and SHED-CM treatment groups, suggesting that SHED improves ALI by paracrine mechanisms. SHED-CM contained multiple therapeutic factors involved in lung-regenerative mechanisms. Importantly, SHED-CM attenuated the BLM-induced pro-inflammatory response and generated an anti-inflammatory/tissue-regenerating environment, accompanied by the induction of anti-inflammatory M2-like lung macrophages. Furthermore, SHED-CM promoted the in vitro differentiation of bone marrow-derived macrophages into M2-like cells, which expressed high levels of Arginase1, CD206 and Ym-1. DISCUSSION: Our results suggest that SHED-secreted factors provide multifaceted therapeutic effects, including a strong M2-inducing activity, for treating BLM-induced ALI. This work may open new avenues for research on stem cell-based ARDS therapies.


Subject(s)
Acute Lung Injury/therapy , Dental Pulp/cytology , Respiratory Distress Syndrome/therapy , Stem Cell Transplantation , Tooth, Deciduous/cytology , Acute Lung Injury/chemically induced , Animals , Arginase/metabolism , Arginase/pharmacology , Bleomycin/pharmacology , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Dental Pulp/metabolism , Female , Humans , Lectins, C-Type/metabolism , Lung/drug effects , Lung/physiology , Macrophages/cytology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mannose-Binding Lectins/pharmacology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/metabolism , Regeneration , Respiratory Distress Syndrome/pathology , Tooth, Deciduous/metabolism
7.
Brain Res ; 1613: 59-72, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25863132

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder caused by the loss of nigrostriatal dopaminergic (DAergic) neurons and the depletion of striatal dopamine. Here we show that DAergic-neuron-like cells could be efficiently induced from stem cells derived from human exfoliated deciduous teeth (SHEDs), and that these induced cells had therapeutic benefits in a 6-OHDA-induced Parkinsonian rat model. In our protocol, EGF and bFGF signaling activated the SHED's expression of proneural genes, Ngn2 and Mash1, and subsequent treatment with brain-derived neurotrophic factor (BDNF) promoted their maturation into DAergic neuron-like SHEDs (dSHEDs). A hypoxic DAergic differentiation protocol improved cell viability and enhanced the expression of multiple neurotrophic factors, including BDNF, GDNF, NT-3, and HGF. Engrafted dSHEDs survived in the striatum of Parkinsonian rats, improved the DA level more efficiently than engrafted undifferentiated SHEDs, and promoted the recovery from neurological deficits. Our findings further suggested that paracrine effects of dSHEDs contributed to neuroprotection against 6-OHDA-induced neurodegeneration and to nigrostriatal tract restoration. In addition, we found that the conditioned medium derived from dSHEDs protected primary neurons against 6-OHDA toxicity and accelerated neurite outgrowth in vitro. Thus, our data suggest that stem cells derived from dental pulp may have therapeutic benefits for PD.


Subject(s)
Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Neural Stem Cells/physiology , Parkinson Disease/surgery , Tooth, Deciduous/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation , Cell Survival , Child , Dopaminergic Neurons/metabolism , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Humans , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Rats , Tooth, Deciduous/drug effects
8.
J Neurosci ; 35(6): 2452-64, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673840

ABSTRACT

Engrafted mesenchymal stem cells from human deciduous dental pulp (SHEDs) support recovery from neural insults via paracrine mechanisms that are poorly understood. Here we show that the conditioned serum-free medium (CM) from SHEDs, administered intrathecally into rat injured spinal cord during the acute postinjury period, caused remarkable functional recovery. The ability of SHED-CM to induce recovery was associated with an immunoregulatory activity that induced anti-inflammatory M2-like macrophages. Secretome analysis of the SHED-CM revealed a previously unrecognized set of inducers for anti-inflammatory M2-like macrophages: monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (ED-Siglec-9). Depleting MCP-1 and ED-Siglec-9 from the SHED-CM prominently reduced its ability to induce M2-like macrophages and to promote functional recovery after spinal cord injury (SCI). The combination of MCP-1 and ED-Siglec-9 synergistically promoted the M2-like differentiation of bone marrow-derived macrophages in vitro, and this effect was abolished by a selective antagonist for CC chemokine receptor 2 (CCR2) or by the genetic knock-out of CCR2. Furthermore, MCP-1 and ED-Siglec-9 administration into the injured spinal cord induced M2-like macrophages and led to a marked recovery of hindlimb locomotor function after SCI. The inhibition of this M2 induction through the inactivation of CCR2 function abolished the therapeutic effects of both SHED-CM and MCP-1/ED-Siglec-9. Macrophages activated by MCP-1 and ED-Siglec-9 extended neurite and suppressed apoptosis of primary cerebellar granule neurons against the neurotoxic effects of chondroitin sulfate proteoglycans. Our data suggest that the unique combination of MCP-1 and ED-Siglec-9 repairs the SCI through anti-inflammatory M2-like macrophage induction.


Subject(s)
Antigens, CD/pharmacology , Chemokine CCL2/pharmacology , Macrophages/drug effects , Sialic Acid Binding Immunoglobulin-like Lectins/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Antigens, CD/metabolism , Blood-Brain Barrier/drug effects , Brain Injuries/drug therapy , Cell Polarity/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Chemokine CCL2/metabolism , Child , Culture Media, Conditioned , Cytokines/metabolism , Dental Pulp/cytology , Dental Pulp/metabolism , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, CCR2/antagonists & inhibitors , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Spinal Cord Injuries/pathology , Tooth, Deciduous
9.
Methods Mol Biol ; 1213: 321-8, 2014.
Article in English | MEDLINE | ID: mdl-25173394

ABSTRACT

Spinal cord injury (SCI) involves concurrent, interacting pathological processes, and requires a multifaceted therapeutic strategy. Stem cell-based transplantation holds great promise as such an approach. We have reported that stem cells derived from human dental pulp have remarkable neuroregenerative activity, and that when transplanted into animal models of SCI, these cells promote functional recovery by inhibiting massive SCI-induced apoptosis, preserving neural fibers and myelin, regenerating transected axons, and replacing damaged cells by differentiating into oligodendrocytes. Here, we introduce some details of our experimental procedures, which may serve as a guide for designing experiments to evaluate the therapeutic benefits of various types of stem cells.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Adult , Animals , Cell Line , Culture Media, Conditioned , Dental Pulp/cytology , Disease Models, Animal , Female , Humans , Rats , Recovery of Function , Spinal Cord Injuries/rehabilitation , Young Adult
10.
Bone ; 61: 82-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24389414

ABSTRACT

Distraction osteogenesis (DO) successfully induces large-scale skeletal tissue regeneration, but it involves an undesirably long treatment period. A high-speed DO mouse model (H-DO) with a distraction speed twice that of a control DO model failed to generate new bone callus in the distraction gap. Here we demonstrate that the local administration of serum-free conditioned medium from human mesenchymal stem cells (MSC-CM) accelerated callus formation in the mouse H-DO model. Secretomic analysis identified factors contained in MSC-CM that recruit murine bone marrow stromal cells (mBMSCs) and endothelial cells/endothelial progenitor cells (EC/EPCs), inhibit inflammation and apoptosis, and promote osteoblast differentiation, angiogenesis, and cell proliferation. Functional assays identified MCP-1/-3 and IL-3/-6 as essential factors in recruiting mBMSCs and EC/EPCs. IL-3/-6 also enhanced the osteogenic differentiation of mBMSCs. MSC-CM that had been depleted of MCP-1/-3 failed to recruit mBMSCs, and consequently failed to promote callus formation. Taken together, our data suggest that MSCs produce a broad repertoire of trophic factors with tissue-regenerative activities that accelerate healing in the DO process.


Subject(s)
Bone and Bones/metabolism , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis, Distraction/methods , Osteogenesis/drug effects , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred ICR , Osteoblasts/cytology , Osteoblasts/drug effects
11.
Neurosci Res ; 78: 16-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24252618

ABSTRACT

Spinal cord injury (SCI) often leads to persistent functional deficits due to the loss of neurons and glia and to limited axonal regeneration after such injury. Recently, three independent groups have reported marked recovery of hindlimb locomotor function after the transplantation of human adult dental pulp stem cells (DPSCs) and stem cells from human exfoliated deciduous teeth (SHEDs) into rats or mice with acute, sub-acute or chronic SCI. This review summarizes the primary characteristics of human dental pulp stem cells and their therapeutic benefits for treating SCI. Experimental data from multiple preclinical studies suggest that pulp stem cells may promote functional recovery after SCI through multifaceted neuro-regenerative activities.


Subject(s)
Dental Pulp/cytology , Nerve Regeneration/physiology , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Stem Cells/physiology , Animals , Humans , Mice , Neurons/physiology , Rats , Stem Cells/cytology
12.
Stroke ; 44(2): 551-4, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23238858

ABSTRACT

BACKGROUND AND PURPOSE: Perinatal hypoxia-ischemia (HI) has high rates of neurological deficits and mortality. So far, no effective treatment for HI brain injury has been developed. In this study, we investigated the therapeutic effects of stem cells from human exfoliated deciduous teeth (SHED) for the treatment of neonatal HI brain injury. METHODS: Unilateral HI was induced in postnatal day 5 (P5) mice. Twenty-four hours later, SHED, human skin fibroblasts, or serum-free conditioned medium derived from these cells was injected into the injured brain. The effects of cell transplantation or conditioned medium injection on the animals' neurological and pathophysiological recovery were evaluated. RESULTS: Transplanted SHED, but not fibroblasts, significantly reduced the HI-induced brain-tissue loss and improved neurological function. SHED also improved the survival of the HI mice. The engrafted SHED rarely differentiated into neural lineages; however, their transplantation inhibited the expression of proinflammatory cytokines, increased the expression of anti-inflammatory ones, and significantly reduced apoptosis. Notably, the intracerebral administration of SHED-conditioned medium also significantly improved the neurological outcome, inhibited apoptosis, and reduced tissue loss. CONCLUSIONS: SHED transplantation into the HI-injured brain resulted in remarkable neurological and pathophysiological recovery. Our findings indicate that paracrine factors derived from SHED support a neuroprotective microenvironment in the HI brain. SHED graft and SHED-conditioned medium may provide a novel neuroprotective therapy for HI.


Subject(s)
Brain Injuries/surgery , Dental Pulp/cytology , Dental Pulp/transplantation , Hypoxia-Ischemia, Brain/surgery , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Brain Injuries/pathology , Cells, Cultured , Humans , Hypoxia-Ischemia, Brain/pathology , Mice
13.
J Clin Invest ; 122(1): 80-90, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22133879

ABSTRACT

Spinal cord injury (SCI) often leads to persistent functional deficits due to loss of neurons and glia and to limited axonal regeneration after injury. Here we report that transplantation of human dental pulp stem cells into the completely transected adult rat spinal cord resulted in marked recovery of hind limb locomotor functions. Transplantation of human bone marrow stromal cells or skin-derived fibroblasts led to substantially less recovery of locomotor function. The human dental pulp stem cells exhibited three major neuroregenerative activities. First, they inhibited the SCI-induced apoptosis of neurons, astrocytes, and oligodendrocytes, which improved the preservation of neuronal filaments and myelin sheaths. Second, they promoted the regeneration of transected axons by directly inhibiting multiple axon growth inhibitors, including chondroitin sulfate proteoglycan and myelin-associated glycoprotein, via paracrine mechanisms. Last, they replaced lost cells by differentiating into mature oligodendrocytes under the extreme conditions of SCI. Our data demonstrate that tooth-derived stem cells may provide therapeutic benefits for treating SCI through both cell-autonomous and paracrine neuroregenerative activities.


Subject(s)
Adult Stem Cells/transplantation , Dental Pulp/cytology , Nerve Regeneration/physiology , Spinal Cord Injuries/therapy , Animals , Apoptosis , Astrocytes/pathology , Cell Differentiation , Cell Survival , Culture Media, Conditioned , Female , Fibroblasts/transplantation , Hindlimb , Humans , Locomotion/physiology , Myelin Sheath/pathology , Neurons/pathology , Oligodendroglia/pathology , Paracrine Communication , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Stromal Cells/transplantation , Transplantation, Heterologous , rho GTP-Binding Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...