Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cell Reports ; 12(2): 274-289, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30661994

ABSTRACT

The hostile environment of an injured spinal cord makes it challenging to achieve higher viability in a grafted tissue-engineered neural network used to reconstruct the spinal cord circuit. Here, we investigate whether cell survival and synaptic transmission within an NT-3 and TRKC gene-overexpressing neural stem cell-derived neural network scaffold (NN) transplanted into transected spinal cord could be promoted by electroacupuncture (EA) through improving the microenvironment. Our results showed that EA facilitated the cell survival, neuronal differentiation, and synapse formation of a transplanted NN. Pseudorabies virus tracing demonstrated that EA strengthened synaptic integration of the transplanted NN with the host neural circuit. The combination therapy also promoted axonal regeneration, spinal conductivity, and functional recovery. The findings highlight EA as a potential and safe supplementary therapeutic strategy to reinforce the survival and synaptogenesis of a transplanted NN as a neuronal relay to bridge the two severed ends of an injured spinal cord.


Subject(s)
Neural Stem Cells/physiology , Neurons/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord/physiology , Animals , Cell Differentiation/physiology , Electroacupuncture/methods , Female , Nerve Regeneration/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Synapses/physiology , Synaptic Transmission/physiology
2.
J Biomed Mater Res A ; 106(8): 2158-2170, 2018 08.
Article in English | MEDLINE | ID: mdl-29577604

ABSTRACT

Spinal cord injury (SCI) normally results in cell death, scarring, cavitation, inhibitory molecules release, etc., which are regarded as a huge obstacle to reconnect the injured neuronal circuits because of the lack of effective stimulus. In this study, a functional gelatin sponge scaffold was used to inhibit local inflammation, enhance nerve fiber regeneration, and improve neural conduction in the canine. This scaffold had good porosity and modified with neurotrophin-3 (NT-3)/fibroin complex, which showed sustained release in vitro. After the scaffold was transplanted into canine spinal cord hemisection model, hindlimb movement, and neural conduction were improved evidently. Migrating host cells, newly formed neurons with associated synaptic structures together with functional blood vessels with intact endothelium in the regenerating tissue were identified. Taken together, the results demonstrated that using bioactive scaffold could establish effective microenvironment stimuli for endogenous regeneration, providing a potential and practical strategy for treatment of spinal cord injury. © 2018 The Authors Journal of Biomedical Materials Research Part A Published by Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2158-2170, 2018.


Subject(s)
Inflammation/pathology , Motor Activity , Nerve Fibers/physiology , Nerve Regeneration , Neurotrophin 3/pharmacology , Spinal Cord Injuries/physiopathology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/pharmacology , Cell Movement/drug effects , Dogs , Evoked Potentials, Motor/drug effects , Female , Fibroins/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Hindlimb/physiopathology , Motor Activity/drug effects , Nerve Fibers/drug effects , Nerve Regeneration/drug effects , Prostheses and Implants , Spinal Cord/blood supply , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology
3.
Biomaterials ; 160: 37-55, 2018 04.
Article in English | MEDLINE | ID: mdl-29353106

ABSTRACT

The functional multipotency enables mesenchymal stem cells (MSCs) promising translational potentials in treating spinal cord injury (SCI). Yet the fate of MSCs grafted into the injured spinal cord has not been fully elucidated even in preclinical studies, rendering concerns of their safety and genuine efficacy. Here we used a rat spinal cord transection model to evaluate the cell fate of allograft bone marrow derived MSCs. With the application of immunosuppressant, donor cells, delivered by biocompatible scaffold, survived up to 8 weeks post-grafting. Discernible tubes formed by MSCs were observed beginning 2 weeks after transplantation and they dominated the morphological features of implanted MSCs at 8 weeks post-grafting. The results of immunocytochemistry and transmission electron microscopy displayed the formation of perineurium-like sheath by donor cells, which, in a manner comparable to the perineurium in peripheral nerve, enwrapped host myelins and axons. The MSC-derived perineurium-like sheath secreted a group of trophic factors and permissive extracellular matrix, and served as a physical and chemical barrier to insulate the inner nerve fibers from ambient oxidative insults by the secretion of soluble antioxidant, superoxide dismutase-3 (SOD3). As a result, many intact regenerating axons were preserved in the injury/graft site following the forming of perineurium-like sheath. A parallel study utilizing a good manufacturing practice (GMP) grade human umbilical cord-derived MSCs or allogenic MSCs in an acute contusive/compressive SCI model exhibited a similar perineurium-like sheath formed by surviving donor cells in rat spinal cord at 3 weeks post-grafting. The present study for the first time provides an unambiguous morphological evidence of perineurium-like sheath formed by transplanted MSCs and a novel therapeutic mechanism of MSCs in treating SCI.


Subject(s)
Mesenchymal Stem Cells , Peripheral Nerves , Tissue Scaffolds , Animals , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Nerve Regeneration , Neuroprotection , Peripheral Nerves/cytology , Peripheral Nerves/physiology , Rats, Sprague-Dawley , Spinal Cord Injuries/therapy , Tissue Engineering
4.
J Biomed Mater Res A ; 104(8): 1902-11, 2016 08.
Article in English | MEDLINE | ID: mdl-26991461

ABSTRACT

Extracellular matrix (ECM) expression is temporally and spatially regulated during the development of stem cells. We reported previously that fibronectin (FN) secreted by bone marrow mesenchymal stem cells (MSCs) was deposited on the surface of gelatin sponge (GS) soon after culture. In this study, we aimed to assess the function of accumulated FN on neuronal differentiating MSCs as induced by Schwann cells (SCs) in three dimensional transwell co-culture system. The expression pattern and amount of FN of differentiating MSCs was examined by immunofluorescence, Western blot and immunoelectron microscopy. The results showed that FN accumulated inside GS scaffold, although its mRNA expression in MSCs was progressively decreased during neural induction. MSC-derived neuron-like cells showed spindle-shaped cell body and long extending processes on FN-decorated scaffold surface. However, after blocking of FN function by application of monoclonal antibodies, neuron-like cells showed flattened cell body with short and thick neurites, together with decreased expression of integrin ß1. In vivo transplantation study revealed that autocrine FN significantly facilitated endogenous nerve fiber regeneration in spinal cord transection model. Taken together, the present results showed that FN secreted by MSCs in the early stage accumulated on the GS scaffold and promoted the neurite elongation of neuronal differentiating MSCs as well as nerve fiber regeneration after spinal cord injury. This suggests that autocrine FN has a dynamic influence on MSCs in a three dimensional culture system and its potential application for treatment of traumatic spinal cord injury. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1902-1911, 2016.


Subject(s)
Autocrine Communication , Cell Differentiation , Fibronectins/pharmacology , Mesenchymal Stem Cells/cytology , Nerve Regeneration/drug effects , Neurites/metabolism , Spinal Cord Injuries/physiopathology , Animals , Cell Adhesion , Cell Differentiation/drug effects , Coculture Techniques , Female , Gelatin/chemistry , Imaging, Three-Dimensional , Integrin beta1/metabolism , Mesenchymal Stem Cells/drug effects , Nerve Fibers/metabolism , Neurites/drug effects , Protein Binding , Rats, Sprague-Dawley , Rats, Transgenic , Spinal Cord Injuries/pathology
5.
Stem Cell Res Ther ; 6: 105, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26012641

ABSTRACT

INTRODUCTION: Severe spinal cord injury often causes temporary or permanent damages in strength, sensation, or autonomic functions below the site of the injury. So far, there is still no effective treatment for spinal cord injury. Mesenchymal stem cells (MSCs) have been used to repair injured spinal cord as an effective strategy. However, the low neural differentiation frequency of MSCs has limited its application. The present study attempted to explore whether the grafted MSC-derived neural-like cells in a gelatin sponge (GS) scaffold could maintain neural features or transdifferentiate into myelin-forming cells in the transected spinal cord. METHODS: We constructed an engineered tissue by co-seeding of MSCs with genetically enhanced expression of neurotrophin-3 (NT-3) and its high-affinity receptor tropomyosin receptor kinase C (TrkC) separately into a three-dimensional GS scaffold to promote the MSCs differentiating into neural-like cells and transplanted it into the gap of a completely transected rat spinal cord. The rats received extensive post-operation care, including cyclosporin A administrated once daily for 2 months. RESULTS: MSCs modified genetically could differentiate into neural-like cells in the MN + MT (NT-3-MSCs + TrKC-MSCs) group 14 days after culture in the GS scaffold. However, after the MSC-derived neural-like cells were transplanted into the injury site of spinal cord, some of them appeared to lose the neural phenotypes and instead transdifferentiated into myelin-forming cells at 8 weeks. In the latter, the MSC-derived myelin-forming cells established myelin sheaths associated with the host regenerating axons. And the injured host neurons were rescued, and axon regeneration was induced by grafted MSCs modified genetically. In addition, the cortical motor evoked potential and hindlimb locomotion were significantly ameliorated in the rat spinal cord transected in the MN + MT group compared with the GS and MSC groups. CONCLUSION: Grafted MSC-derived neural-like cells in the GS scaffold can transdifferentiate into myelin-forming cells in the completely transected rat spinal cord.


Subject(s)
Axons/physiology , Mesenchymal Stem Cells/cytology , Myelin Sheath/metabolism , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Adenoviridae/genetics , Animals , Behavior, Animal , Cell Culture Techniques , Cell Transdifferentiation , Cells, Cultured , Female , Mesenchymal Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptor, trkC/genetics , Receptor, trkC/metabolism , Recovery of Function , Regeneration , Tissue Scaffolds
6.
Biomaterials ; 53: 184-201, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25890718

ABSTRACT

Functional deficits following spinal cord injury (SCI) primarily attribute to loss of neural connectivity. We therefore tested if novel tissue engineering approaches could enable neural network repair that facilitates functional recovery after spinal cord transection (SCT). Rat bone marrow-derived mesenchymal stem cells (MSCs), genetically engineered to overexpress TrkC, receptor of neurotrophin-3 (NT-3), were pre-differentiated into cells carrying neuronal features via co-culture with NT-3 overproducing Schwann cells in 3-dimensional gelatin sponge (GS) scaffold for 14 days in vitro. Intra-GS formation of MSC assemblies emulating neural network (MSC-GS) were verified morphologically via electron microscopy (EM) and functionally by whole-cell patch clamp recording of spontaneous post-synaptic currents. The differentiated MSCs still partially maintained prototypic property with the expression of some mesodermal cytokines. MSC-GS or GS was then grafted acutely into a 2 mm-wide transection gap in the T9-T10 spinal cord segments of adult rats. Eight weeks later, hindlimb function of the MSC-GS-treated SCT rats was significantly improved relative to controls receiving the GS or lesion only as indicated by BBB score. The MSC-GS transplantation also significantly recovered cortical motor evoked potential (CMEP). Histologically, MSC-derived neuron-like cells maintained their synapse-like structures in vivo; they additionally formed similar connections with host neurites (i.e., mostly serotonergic fibers plus a few corticospinal axons; validated by double-labeled immuno-EM). Moreover, motor cortex electrical stimulation triggered c-fos expression in the grafted and lumbar spinal cord cells of the treated rats only. Our data suggest that MSC-derived neuron-like cells resulting from NT-3-TrkC-induced differentiation can partially integrate into transected spinal cord and this strategy should be further investigated for reconstructing disrupted neural circuits.


Subject(s)
Mesenchymal Stem Cells/cytology , Nerve Net , Neurons/cytology , Spinal Cord/surgery , Animals , Coculture Techniques , Rats , Rats, Sprague-Dawley , Rats, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...