Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biomaterials ; 284: 121526, 2022 05.
Article in English | MEDLINE | ID: mdl-35461098

ABSTRACT

Traumatic damage to the spinal cord does not spontaneously heal, often leading to permanent tissue defects. We have shown that injection of imidazole-poly(organophosphazene) hydrogel (I-5) bridges cystic cavities with the newly assembled fibronectin-rich extracellular matrix (ECM). The hydrogel-created ECM contains chondroitin sulfate proteoglycans (CSPGs), collagenous fibrils together with perivascular fibroblasts, and various fibrotic proteins, all of which could hinder axonal growth in the matrix. In an in vitro fibrotic scar model, fibroblasts exhibited enhanced sensitivity to TGF-ß1 when grown on CSPGs. To alleviate the fibrotic microenvironment, the I-5 hydrogel was equipped with an additional function by making a complex with ARSB, a human enzyme degrading CSPGs, via hydrophobic interaction. Delivery of the I-5/ARSB complex significantly diminished the fibrotic ECM components. The complex promoted serotonergic axonal growth into the hydrogel-induced matrix and enhanced serotonergic innervation of the lumbar motor neurons. Regeneration of the propriospinal axons deep into the matrix and to the lumbar spinal cord was robustly increased accompanied by improved locomotor recovery. Therefore, our dual-functional system upgraded the functionality of the hydrogel for spinal cord regeneration by creating ECM to bridge tissue defects and concurrently facilitating axonal connections through the newly assembled ECM.


Subject(s)
N-Acetylgalactosamine-4-Sulfatase , Spinal Cord Injuries , Spinal Cord Regeneration , Animals , Axons/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Delayed-Action Preparations/metabolism , Humans , Hydrogels/chemistry , N-Acetylgalactosamine-4-Sulfatase/metabolism , N-Acetylgalactosamine-4-Sulfatase/therapeutic use , Nerve Regeneration/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord
2.
J Neurosci ; 41(3): 446-460, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33262248

ABSTRACT

Preconditioning peripheral nerve injury enhances the intrinsic growth capacity of DRGs sensory axons by inducing transcriptional upregulation of the regeneration-associated genes (RAGs). However, it is still unclear how preconditioning injury leads to the orchestrated induction of many RAGs. The present study identified Myc proto-oncogene as a transcriptional hub gene to regulate the expression of a distinct subset of RAGs in DRGs following the preconditioning injury. We demonstrated that c-MYC bound to the promoters of certain RAGs, such as Jun, Atf3, and Sprr1a, and that Myc upregulation following SNI preceded that of the RAGs bound by c-MYC. Marked DNA methylation of the Myc exon 3 sequences was implicated in the early transcriptional activation and accompanied by open histone marks. Myc deletion led to a decrease in the injury-induced expression of a distinct subset of RAGs, which were highly overlapped with the list of RAGs that were upregulated by Myc overexpression. Following dorsal hemisection spinal cord injury in female rats, Myc overexpression in DRGs significantly prevented the retraction of the sensory axons in a manner dependent on its downstream RAG, June Our results suggest that Myc plays a critical role in axon regeneration via its transcriptional activity to regulate the expression of a spectrum of downstream RAGs and subsequent effector molecules. Identification of more upstream hub transcription factors and the epigenetic mechanisms specific for individual hub transcription factors would advance our understanding of how the preconditioning injury induces orchestrated upregulation of RAGs.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Animals , Axons/physiology , DNA Methylation , Epigenesis, Genetic/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Neurites , PC12 Cells , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology
3.
Exp Neurobiol ; 29(1): 70-79, 2020 Feb 29.
Article in English | MEDLINE | ID: mdl-32122109

ABSTRACT

Many previous studies have shown reduced glucose uptake in the ischemic brain. In contrast, in a permanent unilateral common carotid artery occlusion (UCCAO) mouse model, our pilot experiments using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) revealed that a subset of mice exhibited conspicuously high uptake of glucose in the ipsilateral hemisphere at 1 week post-occlusion (asymmetric group), whereas other mice showed symmetric uptake in both hemispheres (symmetric group). Thus, we aimed to understand the discrepancy between the two groups. Cerebral blood flow and histological/metabolic changes were analyzed using laser Doppler flowmetry and immunohistochemistry/Western blotting, respectively. Contrary to the increased glucose uptake observed in the ischemic cerebral hemisphere on FDG PET (p<0.001), cerebral blood flow tended to be lower in the asymmetric group than in the symmetric group (right to left ratio [%], 36.4±21.8 vs. 58.0±24.8, p=0.059). Neuronal death was observed only in the ischemic hemisphere of the asymmetric group. In contrast, astrocytes were more activated in the asymmetric group than in the symmetric group (p<0.05). Glucose transporter-1, and monocarboxylate transporter-1 were also upregulated in the asymmetric group, compared with the symmetric group (p<0.05, respectively). These results suggest that the increased FDG uptake was associated with relatively severe ischemia, and glucose transporter-1 upregulation and astrocyte activation. Glucose metabolism may thus be a compensatory mechanism in the moderately severe ischemic brain.

4.
Neurobiol Dis ; 134: 104672, 2020 02.
Article in English | MEDLINE | ID: mdl-31707117

ABSTRACT

Ischemic white matter injuries underlie cognitive decline in the elderly and vascular dementia. Ischemia in the subcortical white matter is caused by chronic reduction of blood flow due to narrowing of small arterioles. However, it remains unclear how chronic ischemia leads to white matter pathology. We aimed to develop an in vitro model of ischemic white matter injury using organotypic slice cultures. Cultured cerebellar slices preserved fully myelinated white matter tracts that were amenable to chronic hypoxic insult. Prolonged hypoxia caused progressive morphological evidence of axonal degeneration with focal constrictions and swellings. In contrast, myelin sheaths and oligodendrocytes exhibited remarkable resilience to hypoxia. The cytoskeletal degradation of axons was accompanied by mitochondrial shortening and lysosomal activation. Multiple pharmacological manipulations revealed that the AMPA glutamate receptor, calpain proteolysis, and lysosomal proteases were independently implicated in hypoxia-induced axonal degeneration in our model. Thus, our in vitro model would be a novel experimental system to explore molecular mechanisms of ischemic white matter injury. Furthermore, we verified that the in vitro assay could be successfully utilized to screen for molecules that can ameliorate hypoxia/ischemia-induced axonal degeneration.


Subject(s)
Axons/pathology , Axons/physiology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , White Matter/pathology , White Matter/physiopathology , Animals , Cell Hypoxia , Cerebellum/pathology , Cerebellum/physiopathology , Lysosomes/physiology , Mice, Inbred C57BL , Myelin Sheath/pathology , Organ Culture Techniques , Proteolysis , Receptors, AMPA/physiology
5.
Exp Neurobiol ; 27(6): 489-507, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30636901

ABSTRACT

Survival and migration of transplanted neural stem cells (NSCs) are prerequisites for therapeutic benefits in spinal cord injury. We have shown that survival of NSC grafts declines after transplantation into the injured spinal cord, and that combining treadmill training (TMT) enhances NSC survival via insulin-like growth factor-1 (IGF-1). Here, we aimed to obtain genetic evidence that IGF-1 signaling in the transplanted NSCs determines the beneficial effects of TMT. We transplanted NSCs heterozygous (+/-) for Igf1r, the gene encoding IGF-1 receptor, into the mouse spinal cord after injury, with or without combining TMT. We analyzed the influence of genotype and TMT on locomotor recovery and survival and migration of NSC grafts. In vitro experiments were performed to examine the potential roles of IGF-1 signaling in the migratory ability of NSCs. Mice receiving +/- NSC grafts showed impaired locomotor recovery compared with those receiving wild-type (+/+) NSCs. Locomotor improvement by TMT was more pronounced with +/+ grafts. Deficiency of one allele of Igf1r significantly reduced survival and migration of the transplanted NSCs. Although TMT did not significantly influence NSC survival, it substantially enhanced the extent of migration for only +/+ NSCs. Cultured neurospheres exhibited dynamic motility with cytoplasmic protrusions, which was regulated by IGF-1 signaling. IGF-1 signaling in transplanted NSCs may be essential in regulating their survival and migration. Furthermore, TMT may promote NSC graft-mediated locomotor recovery via activation of IGF-1 signaling in transplanted NSCs. Dynamic NSC motility via IGF-1 signaling may be the cellular basis for the TMT-induced enhancement of migration.

6.
Nat Commun ; 8(1): 533, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912446

ABSTRACT

The cystic cavity that develops following injuries to brain or spinal cord is a major obstacle for tissue repair in central nervous system (CNS). Here we report that injection of imidazole-poly(organophosphazenes) (I-5), a hydrogel with thermosensitive sol-gel transition behavior, almost completely eliminates cystic cavities in a clinically relevant rat spinal cord injury model. Cystic cavities are bridged by fibronectin-rich extracellular matrix. The fibrotic extracellular matrix remodeling is mediated by matrix metalloproteinase-9 expressed in macrophages within the fibrotic extracellular matrix. A poly(organophosphazenes) hydrogel lacking the imidazole moiety, which physically interacts with macrophages via histamine receptors, exhibits substantially diminished bridging effects. I-5 injection improves coordinated locomotion, and this functional recovery is accompanied by preservation of myelinated white matter and motor neurons and an increase in axonal reinnervation of the lumbar motor neurons. Our study demonstrates that dynamic interactions between inflammatory cells and injectable biomaterials can induce beneficial extracellular matrix remodeling to stimulate tissue repair following CNS injuries.The cystic cavity that develops following injuries to brain or spinal cord is a major obstacle. Here the authors show an injection of imidazole poly(organophosphazenes), a hydrogel with thermosensitive sol-gel transition behavior, almost completely eliminates cystic cavities in a clinically relevant rat spinal cord injury model.


Subject(s)
Extracellular Matrix/physiology , Hydrogels/administration & dosage , Regeneration/physiology , Spinal Cord Injuries/therapy , Animals , Female , Fibronectins/metabolism , Hydrogels/chemistry , Imidazoles/chemical synthesis , Imidazoles/chemistry , Macrophages/physiology , Matrix Metalloproteinase 9/genetics , Mice , NIH 3T3 Cells , Polymers/chemical synthesis , Polymers/chemistry , Rats, Sprague-Dawley , Spinal Cord/physiology , Spinal Cord Injuries/pathology
8.
J Neurosci ; 35(48): 15934-47, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26631474

ABSTRACT

CNS neurons in adult mammals do not spontaneously regenerate axons after spinal cord injury. Preconditioning peripheral nerve injury allows the dorsal root ganglia (DRG) sensory axons to regenerate beyond the injury site by promoting expression of regeneration-associated genes. We have previously shown that peripheral nerve injury increases the number of macrophages in the DRGs and that the activated macrophages are critical to the enhancement of intrinsic regeneration capacity. The present study identifies a novel chemokine signal mediated by CCL2 that links regenerating neurons with proregenerative macrophage activation. Neutralization of CCL2 abolished the neurite outgrowth activity of conditioned medium obtained from neuron-macrophage cocultures treated with cAMP. The neuron-macrophage interactions that produced outgrowth-promoting conditioned medium required CCL2 in neurons and CCR2/CCR4 in macrophages. The conditioning effects were abolished in CCL2-deficient mice at 3 and 7 d after sciatic nerve injury, but CCL2 was dispensable for the initial growth response and upregulation of GAP-43 at the 1 d time point. Intraganglionic injection of CCL2 mimicked conditioning injury by mobilizing M2-like macrophages. Finally, overexpression of CCL2 in DRGs promoted sensory axon regeneration in a rat spinal cord injury model without harmful side effects. Our data suggest that CCL2-mediated neuron-macrophage interaction plays a critical role for amplification and maintenance of enhanced regenerative capacity by preconditioning peripheral nerve injury. Manipulation of chemokine signaling mediating neuron-macrophage interactions may represent a novel therapeutic approach to promote axon regeneration after CNS injury.


Subject(s)
Chemokine CCL2/metabolism , Macrophages/physiology , Nerve Regeneration/genetics , Neurons/physiology , Peripheral Nerve Injuries/physiopathology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Cholera Toxin/metabolism , Coculture Techniques , Dependovirus/genetics , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Neurites/physiology , Neurons/cytology , Pain Measurement , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
9.
J Neurosci ; 34(38): 12788-800, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232115

ABSTRACT

Combining cell transplantation with activity-based rehabilitation is a promising therapeutic approach for spinal cord repair. The present study was designed to investigate potential interactions between the transplantation (TP) of neural stem cells (NSCs) obtained at embryonic day 14 and treadmill training (TMT) in promoting locomotor recovery and structural repair in rat contusive injury model. Combination of TMT with NSC TP at 1 week after injury synergistically improved locomotor function. We report here that combining TMT increased the survival of grafted NSCs by >3-fold and >5-fold at 3 and 9 weeks after injury, respectively. The number of surviving NSCs was significantly correlated with the extent of locomotor recovery. NSCs grafted into the injured spinal cord were under cellular stresses induced by reactive nitrogen or oxygen species, which were markedly attenuated by TMT. TMT increased the concentration of insulin-like growth factor-1 (IGF-1) in the CSF. Intrathecal infusion of neutralizing IGF-1 antibodies, but not antibodies against either BDNF or Neurotrophin-3 (NT-3), abolished the enhanced survival of NSC grafts by TMT. The combination of TP and TMT also resulted in tissue sparing, increased myelination, and restoration of serotonergic fiber innervation to the lumbar spinal cord to a larger extent than that induced by either TP or TMT alone. Therefore, we have discovered unanticipated beneficial effects of TMT in modulating the survival of grafted NSCs via IGF-1. Our study identifies a novel neurobiological basis for complementing NSC-based spinal cord repair with activity-based neurorehabilitative approaches.


Subject(s)
Insulin-Like Growth Factor I/physiology , Motor Activity/physiology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Signal Transduction , Spinal Cord Injuries/rehabilitation , Spinal Cord Injuries/therapy , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Brain-Derived Neurotrophic Factor/immunology , Cell Survival/immunology , Cell Survival/physiology , Combined Modality Therapy/methods , Female , Injections, Spinal , Insulin-Like Growth Factor I/immunology , Insulin-Like Growth Factor I/metabolism , Lumbosacral Region/innervation , Myelin Sheath/metabolism , Neurotrophin 3/immunology , Rats , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Recovery of Function/physiology , Serotonergic Neurons/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Spinal Cord Regeneration/immunology , Spinal Cord Regeneration/physiology
10.
PLoS One ; 9(2): e88215, 2014.
Article in English | MEDLINE | ID: mdl-24520355

ABSTRACT

Traumatic spinal cord injury (SCI) often leads to debilitating loss of locomotor function. Neuroplasticity of spinal circuitry underlies some functional recovery and therefore represents a therapeutic target to improve locomotor function following SCI. However, the cellular and molecular mechanisms mediating neuroplasticity below the lesion level are not fully understood. The present study performed a gene expression profiling in the rat lumbar spinal cord at 1 and 3 weeks after contusive SCI at T9. Another group of rats received treadmill locomotor training (TMT) until 3 weeks, and gene expression profiles were compared between animals with and without TMT. Microarray analysis showed that many inflammation-related genes were robustly upregulated in the lumbar spinal cord at both 1 and 3 weeks after thoracic injury. Notably, several components involved in an early complement activation pathway were concurrently upregulated. In line with the microarray finding, the number of microglia substantially increased not only in the white matter but also in the gray matter. C3 and complement receptor 3 were intensely expressed in the ventral horn after injury. Furthermore, synaptic puncta near ventral motor neurons were frequently colocalized with microglia after injury, implicating complement activation and microglial cells in synaptic remodeling in the lumbar locomotor circuitry after SCI. Interestingly, TMT did not influence the injury-induced upregulation of inflammation-related genes. Instead, TMT restored pre-injury expression patterns of several genes that were downregulated by injury. Notably, TMT increased the expression of genes involved in neuroplasticity (Arc, Nrcam) and angiogenesis (Adam8, Tie1), suggesting that TMT may improve locomotor function in part by promoting neurovascular remodeling in the lumbar motor circuitry.


Subject(s)
Lumbar Vertebrae/pathology , Motor Activity/genetics , Physical Conditioning, Animal , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Spinal Cord/pathology , Thoracic Injuries/genetics , Animals , Down-Regulation/genetics , Female , Gene Expression Profiling , Inflammation/genetics , Inflammation/pathology , Lumbar Vertebrae/physiopathology , Microglia/metabolism , Microglia/pathology , Oligonucleotide Array Sequence Analysis , Protein Interaction Maps/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function/genetics , Reproducibility of Results , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Thoracic Injuries/pathology , Thoracic Injuries/physiopathology , Up-Regulation/genetics
11.
J Neurosci ; 33(38): 15095-108, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048840

ABSTRACT

Although the central branches of the dorsal root ganglion (DRG) sensory neurons do not spontaneously regenerate, a conditioning peripheral injury can promote their regeneration. A potential role of macrophages in axonal regeneration was proposed, but it has not been critically addressed whether macrophages play an essential role in the conditioning injury model. After sciatic nerve injury (SNI) in rats, the number of macrophages in DRGs gradually increased by day 7. The increase persisted up to 28 d and was accompanied by upregulation of inflammatory mediators, including oncomodulin. A macrophage deactivator, minocycline, reduced the macrophage number and expressions of the inflammatory mediators. Molecular signatures of conditioning effects were abrogated by minocycline, and enhanced regenerative capacity was substantially attenuated both in vitro and in vivo. Delayed minocycline infusion abrogated the SNI-induced long-lasting heightened neurite outgrowth potential, indicating a role for macrophages in the maintenance of regenerative capacity. Intraganglionic cAMP injection also resulted in an increase in macrophages, and minocycline abolished the cAMP effect on neurite outgrowth. However, conditioned media (CM) from macrophages treated with cAMP did not exhibit neurite growth-promoting activity. In contrast, CM from neuron-macrophage cocultures treated with cAMP promoted neurite outgrowth greatly, highlighting a requirement for neuron-macrophage interactions for the induction of a proregenerative macrophage phenotype. The growth-promoting activity in the CM was profoundly attenuated by an oncomodulin neutralizing antibody. These results suggest that the neuron-macrophage interactions involved in eliciting a proregenerative phenotype in macrophages may be a novel target to induce long-lasting regenerative processes after axonal injuries in the CNS.


Subject(s)
Ganglia, Spinal/pathology , Macrophages/physiology , Nerve Regeneration/physiology , Sciatic Neuropathy/pathology , Sensory Receptor Cells/physiology , Analysis of Variance , Animals , Axons/physiology , Calcium-Binding Proteins/metabolism , Cell Separation , Cells, Cultured , Cholera Toxin/metabolism , Coculture Techniques , Cyclic AMP/pharmacology , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein , Macrophages/drug effects , Microfilament Proteins/metabolism , Minocycline/pharmacology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/drug effects
12.
J Nanosci Nanotechnol ; 13(8): 5513-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23882787

ABSTRACT

Real-time pulse measurements of nano-scale field effect transistors (FETs) are reported. We demonstrate the direct monitoring of the real-time current of bottom-up assembled silicon nanowire FET and top-down fabricated gate-all-around silicon nanowire FET, both with the diameter of approximately 50 nm. We demonstrate that the displacement current can be cancelled out from the measured pulse responses. On the other hand, the displacement current also can be utilized to obtain the coupling capacitance between the gate and source of the FETs.

13.
J Neuroinflammation ; 9: 100, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632146

ABSTRACT

BACKGROUND: Traumatic spinal cord injury (SCI) causes acute neuronal death followed by delayed secondary neuronal damage. However, little is known about how microenvironment regulating cells such as microglia, astrocytes, and blood inflammatory cells behave in early SCI states and how they contribute to delayed neuronal death. METHODS: We analyzed the behavior of neurons and microenvironment regulating cells using a contusion-induced SCI model, examining early (3-6 h) to late times (14 d) after the injury. RESULTS: At the penumbra region close to the damaged core (P1) neurons and astrocytes underwent death in a similar spatial and temporal pattern: both neurons and astrocytes died in the medial and ventral regions of the gray matter between 12 to 24 h after SCI. Furthermore, mRNA and protein levels of transporters of glutamate (GLT-1) and potassium (Kir4.1), functional markers of astrocytes, decreased at about the times that delayed neuronal death occurred. However, at P1 region, ramified Iba-1+ resident microglia died earlier (3 to 6 h) than neurons (12 to 24 h), and at the penumbra region farther from the damaged core (P2), neurons were healthy where microglia were morphologically activated. In addition, round Iba-1/CD45-double positive monocyte-like cells appeared after neurons had died, and expressed phagocytic markers, including mannose receptors, but rarely expressed proinflammatory mediators. CONCLUSION: Loss of astrocyte function may be more critical for delayed neuronal death than microglial activation and monocyte infiltration.


Subject(s)
Astrocytes/pathology , Contusions/pathology , Disease Progression , Nerve Degeneration/pathology , Neurons/pathology , Spinal Cord Injuries/pathology , Animals , Astrocytes/metabolism , Calcium-Binding Proteins/biosynthesis , Contusions/metabolism , Female , Microfilament Proteins/biosynthesis , Nerve Degeneration/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Time Factors
14.
Cytotherapy ; 12(6): 841-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20629485

ABSTRACT

BACKGROUND AIMS: Combinatorial approaches employing diverse therapeutic modalities are required for clinically relevant repair of injured spinal cord in human patients. Before translation into the clinic, the feasibility and therapeutic potential of such combinatorial strategies in larger animal species need to be examined. METHODS: The present study tested the feasibility of implanting polymer scaffolds via neural stem cell (NSC) delivery in a canine spinal cord injury (SCI) model. The poly(lactic-co-glycolic acid) (PLGA) scaffolds seeded with human NSC were implanted into hemisected canine spinal cord. RESULTS: The PLGA scaffolds bridged tissue defects and were nicely integrated with residual canine spinal cord tissue. Grafted NSC survived the implantation procedure and showed migratory behavior to residual spinal cord tissue. Ectopic expression of a therapeutic neurotrophin-3 gene was also possible in NSC seeded within the PLGA scaffolds. CONCLUSIONS: Our description of a canine SCI model would be a valuable resource for pre-clinical trials of combinatorial strategies in larger animal models.


Subject(s)
Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Tissue Scaffolds/statistics & numerical data , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Cell Movement , Disease Models, Animal , Dogs , Feasibility Studies , Graft Survival , Humans , Lactic Acid/chemistry , Lactic Acid/metabolism , Neural Stem Cells/pathology , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Polyglycolic Acid/chemistry , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Tissue Engineering , Tissue Scaffolds/chemistry , Transplantation, Heterologous
15.
PLoS One ; 4(3): e4987, 2009.
Article in English | MEDLINE | ID: mdl-19319198

ABSTRACT

The present study was undertaken to examine multifaceted therapeutic effects of vascular endothelial growth factor (VEGF) in a rat spinal cord injury (SCI) model, focusing on its capability to stimulate proliferation of endogenous glial progenitor cells. Neural stem cells (NSCs) can be genetically modified to efficiently transfer therapeutic genes to diseased CNS. We adopted an ex vivo approach using immortalized human NSC line (F3 cells) to achieve stable and robust expression of VEGF in the injured spinal cord. Transplantation of NSCs retrovirally transduced to overexpress VEGF (F3.VEGF cells) at 7 days after contusive SCI markedly elevated the amount of VEGF in the injured spinal cord tissue compared to injection of PBS or F3 cells without VEGF. Concomitantly, phosphorylation of VEGF receptor flk-1 increased in F3.VEGF group. Stereological counting of BrdU+ cells revealed that transplantation of F3.VEGF significantly enhanced cellular proliferation at 2 weeks after SCI. The number of proliferating NG2+ glial progenitor cells (NG2+/BrdU+) was also increased by F3.VEGF. Furthermore, transplantation of F3.VEGF increased the number of early proliferating cells that differentiated into mature oligodendrocytes, but not astrocytes, at 6 weeks after SCI. F3.VEGF treatment also increased the density of blood vessels in the injured spinal cord and enhanced tissue sparing. These anatomical results were accompanied by improved BBB locomotor scores. The multifaceted effects of VEGF on endogenous gliogenesis, angiogenesis, and tissue sparing could be utilized to improve functional outcomes following SCI.


Subject(s)
Neovascularization, Physiologic , Neuroglia/cytology , Neurons/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Cell Proliferation , Humans , Neurons/metabolism , Rats , Treatment Outcome
16.
Cell Transplant ; 16(4): 355-64, 2007.
Article in English | MEDLINE | ID: mdl-17658126

ABSTRACT

Traumatic injuries to the spinal cord lead to severe and permanent neurological deficits. Although no effective therapeutic option is currently available, recent animal studies have shown that cellular transplantation strategies hold promise to enhance functional recovery after spinal cord injury (SCI). This review is to analyze the experiments where transplantation of stem/progenitor cells produced successful functional outcome in animal models of SCI. There is no consensus yet on what kind of stem/progenitor cells is an ideal source for cellular grafts. Three kinds of stem/progenitor cells have been utilized in cell therapy in animal models of SCI: embryonic stem cells, bone marrow mesenchymal stem cells, and neural stem cells. Neural stem cells or fate-restricted neuronal or glial progenitor cells were preferably used because they have clear capacity to become neurons or glial cells after transplantation into the injured spinal cord. At least a part of functional deficits after SCI is attributable to chronic progressive demyelination. Therefore, several studies transplanted glial-restricted progenitors or oligodendrocyte precursors to target the demyelination process. Directed differentiation of stem/progenitor cells to oligodendrocyte lineage prior to transplantation or modulation of microenvironment in the injured spinal cord to promote oligodendroglial differentiation seems to be an effective strategy to increase the extent of remyelination. Transplanted stem/progenitor cells can also contribute to promoting axonal regeneration by functioning as cellular scaffolds for growing axons. Combinatorial approaches using polymer scaffolds to fill the lesion cavity or introducing regeneration-promoting genes will greatly increase the efficacy of cellular transplantation strategies for SCI.


Subject(s)
Spinal Cord Injuries/therapy , Stem Cell Transplantation/methods , Animals , Bone Marrow Transplantation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation , Neurons/transplantation
17.
J Clin Neurol ; 3(4): 181-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-19513129

ABSTRACT

BACKGROUND AND PURPOSE: N-methyl-D-aspartate (NMDA)-mediated neurotoxicity and oxidative stress have been implicated in the etiology of amyotrophic lateral sclerosis (ALS). Memantine is a low-affinity, noncompetitive NMDA receptor antagonist that may protect against motor neuron degeneration. METHODS: Thirty transgenic mice expressing the G93A SOD1 mutation were randomly divided into control, low-dose memantine (30 mg/kg/day), and high-dose memantine (90 mg/kg/day) groups, with memantine supplied daily with drinking water beginning at 75 days of age. Body weight, survival, and behavioral performances including a rotarod test, paw grip endurance, and hindlimb extension reflex were assessed in the control and memantine-diet groups. RESULTS: Clinical symptoms were evident in the G93A transgenic mice by 11 weeks of age. Memantine was tolerated well. Compared to control, mice treated with memantine performed better in the rotarod test and hindlimb extension reflex. Moreover, low-dose memantine treatment significantly prolonged the survival of the transgenic mice relative to control mice (141 vs 134 days, p<0.05). CONCLUSIONS: These findings suggest that memantine, even when administered at the time of symptom onset, has beneficial effects on patients with ALS.

SELECTION OF CITATIONS
SEARCH DETAIL
...