Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 13(36): 42429-42441, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34472351

ABSTRACT

A critical challenge in many pharmaceutical fields is developing versatile adjuvant devices that can reduce the off-target delivery of therapeutic materials to target lesions. Herein, a biphasic hybrid fibrous system that can manipulate the spatial and temporal delivery of various therapeutic agents to target lesions by integrating multiple distinct systems and technologies such as fluffy coaxial electrospun polycaprolactone (PCL)/polystyrene (PS) fibers, cyclohexane-mediated leaching to remove PS layers selectively, amine display on PCL fibers, conjugation of naturally occurring adhesive gallol molecules onto hyaluronic acid (HA-g), and electrostatically complexing the aminated PCL fibers with the gallol-conjugated HA. In the context of "paintable" systems on target lesions, the resulting system is called a PAINT matrix (abbreviated according to the initial letter of its features: pastable, adhesive, injectable, nanofibrous, and tunable). Its viscoelastic property, which was attributed by coalescing aminated PCL fibers with viscous HA-g, enabled it to be noninvasively injected and fit into any cavity in the body with various morphologies, manually pasted on tissue surfaces, and adhered onto moisture-rich surfaces to ensure the secure delivery of therapeutics toward the target lesions. The PAINT matrix efficiently supplied immunomodulatory human neural stem cells (hNSCs) at rat hemisectioned spinal cord injury (SCI) sites and promoted both locomotive and sensory recovery in SCI models, presumably by protecting hNSCs against host immunosurveillance. The PAINT matrix will be broadly utilized for efficiently delivering therapeutics to difficult-to-reach target lesions by direct infusion or conventional biomaterial-mediated approaches due to their locations, wet surfaces, or complicated ambient environments.


Subject(s)
Adhesives/chemistry , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Animals , HEK293 Cells , Humans , Hyaluronic Acid/chemistry , Male , Nanofibers/chemistry , Phenols/chemistry , Polyesters/chemistry , Rats, Sprague-Dawley , Viscoelastic Substances/chemistry
2.
Yonsei Med J ; 61(6): 492-505, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32469173

ABSTRACT

PURPOSE: To elucidate the brain's intrinsic response to injury, we tracked the response of neural stem/progenitor cells (NSPCs) located in ventricular-subventricular zone (V-SVZ) to hypoxic-ischemic brain injury (HI). We also evaluated whether transduction of V-SVZ NSPCs with neurogenic factor NeuroD1 could enhance their neurogenesis in HI. MATERIALS AND METHODS: Unilateral HI was induced in ICR neonatal mice. To label proliferative V-SVZ NSPCs in response to HI, bromodeoxyuridine (BrdU) and retroviral particles encoding LacZ or NeuroD1/GFP were injected. The cellular responses of NSPCs were analyzed by immunohistochemistry. RESULTS: Unilateral HI increased the number of BrdU+ newly-born cells in the V-SVZ ipsilateral to the lesion while injury reduced the number of newly-born cells reaching the ipsilateral olfactory bulb, which is the programmed destination of migratory V-SVZ NSPCs in the intact brain. These newly-born cells were directed from this pathway towards the lesions. HI significantly increased the number of newly-born cells in the cortex and striatum by the altered migration of V-SVZ cells. Many of these newly-born cells differentiated into active neurons and glia. LacZ-expressing V-SVZ NSPCs also showed extensive migration towards the non-neurogenic regions ipsilateral to the lesion, and expressed the neuronal marker NeuN. NeuroD1+/GFP+ V-SVZ NSPCs almost differentiated into neurons in the peri-infarct regions. CONCLUSION: HI promotes the establishment of a substantial number of new neurons in non-neurogenic regions, suggesting intrinsic repair mechanisms of the brain, by controlling the behavior of endogenous NSPCs. The activation of NeuroD1 expression may improve the therapeutic potential of endogenous NSPCs by increasing their neuronal differentiation in HI.


Subject(s)
Hypoxia-Ischemia, Brain/therapy , Lateral Ventricles/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Hypoxia-Ischemia, Brain/pathology , Mice, Inbred ICR , Nerve Tissue Proteins/metabolism , Nestin/metabolism
3.
Cells ; 9(5)2020 05 11.
Article in English | MEDLINE | ID: mdl-32403417

ABSTRACT

Neural progenitor cells (NPCs) therapy offers great promise in hypoxic-ischemic (HI) brain injury. However, the poor survival of implanted NPCs in the HI host environment limits their therapeutic effects. Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine that is induced in response to a variety of pathological processes including inflammation and immunity. On the other hand, TNF-α has protective effects on cell apoptosis and death and affects the differentiation, proliferation, and survival of neural stem/progenitor cells in the brain. The present study investigated whether TNF-α pretreatment on human NPCs (hNPCs) enhances the effectiveness of cell transplantation therapy under ischemic brain. Fetal brain tissue-derived hNPCs were pretreated with TNF-α before being used in vitro experiments or transplantation. TNF-α significantly increased expression of cIAP2, and the use of short hairpin RNA-mediated knockdown of cIAP2 demonstrated that cIAP2 protected hNPCs against HI-induced cytotoxicity. In addition, pretreatment of hNPCs with TNF-α mediated neuroprotection by altering microglia polarization via increased expression of CX3CL1 and by enhancing expression of neurotrophic factors. Furthermore, transplantation of TNF-α-treated hNPCs reduced infarct volume and improved neurological functions in comparison with non-pretreated hNPCs or vehicle. These findings show that TNF-α pretreatment, which protects hNPCs from HI-injured brain-induced apoptosis and increases neuroprotection, is a simple and safe approach to improve the survival of transplanted hNPCs and the therapeutic efficacy of hNPCs in HI brain injury.


Subject(s)
Brain Injuries/therapy , Hypoxia-Ischemia, Brain/therapy , Neural Stem Cells/transplantation , Tumor Necrosis Factor-alpha/pharmacology , Animals , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Behavior, Animal/drug effects , Brain Injuries/complications , Brain Injuries/pathology , Caspase 3/metabolism , Cell Line , Cell Polarity/drug effects , Cell Survival/drug effects , Chemokine CX3CL1/metabolism , Culture Media, Conditioned/pharmacology , Glutamic Acid/toxicity , Humans , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/pathology , Mice, Inbred ICR , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Growth Factors/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotection/drug effects , Phenotype , Stress, Physiological/drug effects , Up-Regulation/drug effects
4.
Exp Neurobiol ; 28(6): 679-696, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31902156

ABSTRACT

Spinal cord injury (SCI) causes axonal damage and demyelination, neural cell death, and comprehensive tissue loss, resulting in devastating neurological dysfunction. Neural stem/progenitor cell (NSPCs) transplantation provides therapeutic benefits for neural repair in SCI, and glial cell linederived neurotrophic factor (GDNF) has been uncovered to have capability of stimulating axonal regeneration and remyelination after SCI. In this study, to evaluate whether GDNF would augment therapeutic effects of NSPCs for SCI, GDNF-encoding or mock adenoviral vector-transduced human NSPCs (GDNF-or Mock-hNSPCs) were transplanted into the injured thoracic spinal cords of rats at 7 days after SCI. Grafted GDNFhNSPCs showed robust engraftment, long-term survival, an extensive distribution, and increased differentiation into neurons and oligodendroglial cells. Compared with Mock-hNSPC- and vehicle-injected groups, transplantation of GDNF-hNSPCs significantly reduced lesion volume and glial scar formation, promoted neurite outgrowth, axonal regeneration and myelination, increased Schwann cell migration that contributed to the myelin repair, and improved locomotor recovery. In addition, tract tracing demonstrated that transplantation of GDNF-hNSPCs reduced significantly axonal dieback of the dorsal corticospinal tract (dCST), and increased the levels of dCST collaterals, propriospinal neurons (PSNs), and contacts between dCST collaterals and PSNs in the cervical enlargement over that of the controls. Finally grafted GDNF-hNSPCs substantially reversed the increased expression of voltage-gated sodium channels and neuropeptide Y, and elevated expression of GABA in the injured spinal cord, which are involved in the attenuation of neuropathic pain after SCI. These findings suggest that implantation of GDNF-hNSPCs enhances therapeutic efficiency of hNSPCs-based cell therapy for SCI.

5.
Exp Mol Med ; 50(4): 1-18, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29674624

ABSTRACT

Hypoxic-ischemic (HI) brain injury and spinal cord injury (SCI) lead to extensive tissue loss and axonal degeneration. The combined application of the polymer scaffold and neural progenitor cells (NPCs) has been reported to enhance neural repair, protection and regeneration through multiple modes of action following neural injury. This study investigated the reparative ability and therapeutic potentials of biological bridges composed of human fetal brain-derived NPCs seeded upon poly(glycolic acid)-based scaffold implanted into the infarction cavity of a neonatal HI brain injury or the hemisection cavity in an adult SCI. Implantation of human NPC (hNPC)-scaffold complex reduced the lesion volume, induced survival, engraftment, and differentiation of grafted cells, increased neovascularization, inhibited glial scar formation, altered the microglial/macrophage response, promoted neurite outgrowth and axonal extension within the lesion site, and facilitated the connection of damaged neural circuits. Tract tracing demonstrated that hNPC-scaffold grafts appear to reform the connections between neurons and their targets in both cerebral hemispheres in HI brain injury and protect some injured corticospinal fibers in SCI. Finally, the hNPC-scaffold complex grafts significantly improved motosensory function and attenuated neuropathic pain over that of the controls. These findings suggest that, with further investigation, this optimized multidisciplinary approach of combining hNPCs with biomaterial scaffolds provides a more versatile treatment for brain injury and SCI.


Subject(s)
Brain Injuries/therapy , Cells, Immobilized/transplantation , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Tissue Scaffolds/chemistry , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Heterografts , Humans , Mice , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
6.
Transl Res ; 183: 121-136.e9, 2017 05.
Article in English | MEDLINE | ID: mdl-28081931

ABSTRACT

Neonatal hypoxic-ischemic (HI) brain injury leads to high mortality and neurodevelopmental disabilities. Multipotent neural progenitor cells (NPCs) with self-renewing capacity have the potential to reduce neuronal loss and improve the compromised environment in the HI brain injury. However, the therapeutic efficacy of neuronal-committed progenitor cells and the underlying mechanisms of recovery are not yet fully understood. Therefore, this study investigated the regenerative ability and action mechanisms of neuronally committed human NPCs (hNPCs) transduced with neurogenin-2 (NEUROG2) in neonatal HI brain injury. NEUROG2- or green fluorescent protein (GFP)-encoding adenoviral vector-transduced hNPCs (NEUROG2- or GFP-NPCs) were transplanted into neonatal mouse brains with HI injury. Grafted NEUROG2-NPCs showed robust dispersion and engraftment, prolonged survival, and neuronal differentiation in HI brain injury. NEUROG2-NPCs significantly improved neurological behaviors, decreased cellular apoptosis, and increased the neurite outgrowth and axonal sprouting in HI brain injury. In contrast, GFP-NPC grafts moderately enhanced axonal extension with limited behavioral recovery. Notably, NEUROG2-NPCs showed increased secretion of multiple factors, such as nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3 (NTF3), fibroblast growth factor 9 (FGF9), ciliary neurotrophic factor (CNTF), and thrombospondins 1 and 2 (THBS 1/2), which promoted SH-SY5Y neuroblastoma cell survival and neurite outgrowth. Thus, we postulate that NEUROG2-expressing human NPCs facilitate functional recovery after neonatal HI brain injury via their ability to secrete multiple factors that enhance neuronal survival and neuroplasticity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Injuries/therapy , Hypoxia-Ischemia, Brain/therapy , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line, Tumor , Cell Transplantation , Gene Expression Regulation , Humans , Mice , Multipotent Stem Cells , Nerve Tissue Proteins/genetics , Neurons/physiology , Tissue Culture Techniques
7.
Neural Plast ; 2015: 630932, 2015.
Article in English | MEDLINE | ID: mdl-26568892

ABSTRACT

In a phase I/IIa open-label and nonrandomized controlled clinical trial, we sought to assess the safety and neurological effects of human neural stem/progenitor cells (hNSPCs) transplanted into the injured cord after traumatic cervical spinal cord injury (SCI). Of 19 treated subjects, 17 were sensorimotor complete and 2 were motor complete and sensory incomplete. hNSPCs derived from the fetal telencephalon were grown as neurospheres and transplanted into the cord. In the control group, who did not receive cell implantation but were otherwise closely matched with the transplantation group, 15 patients with traumatic cervical SCI were included. At 1 year after cell transplantation, there was no evidence of cord damage, syrinx or tumor formation, neurological deterioration, and exacerbating neuropathic pain or spasticity. The American Spinal Injury Association Impairment Scale (AIS) grade improved in 5 of 19 transplanted patients, 2 (A → C), 1 (A → B), and 2 (B → D), whereas only one patient in the control group showed improvement (A → B). Improvements included increased motor scores, recovery of motor levels, and responses to electrophysiological studies in the transplantation group. Therefore, the transplantation of hNSPCs into cervical SCI is safe and well-tolerated and is of modest neurological benefit up to 1 year after transplants. This trial is registered with Clinical Research Information Service (CRIS), Registration Number: KCT0000879.


Subject(s)
Cervical Cord/injuries , Fetal Stem Cells/transplantation , Neural Stem Cells/transplantation , Spinal Cord Injuries/rehabilitation , Spinal Cord Injuries/therapy , Stem Cell Transplantation/adverse effects , Adolescent , Adult , Female , Humans , Lower Extremity/innervation , Lower Extremity/physiopathology , Male , Middle Aged , Motor Activity , Muscle Spasticity , Neural Conduction , Pain Measurement , Spinal Cord Injuries/pathology , Treatment Outcome , Upper Extremity/innervation , Upper Extremity/physiopathology , Young Adult
8.
EMBO J ; 34(23): 2971-83, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26497893

ABSTRACT

The generation of patient-specific oligodendrocyte progenitor cells (OPCs) holds great potential as an expandable cell source for cell replacement therapy as well as drug screening in spinal cord injury or demyelinating diseases. Here, we demonstrate that induced OPCs (iOPCs) can be directly derived from adult mouse fibroblasts by Oct4-mediated direct reprogramming, using anchorage-independent growth to ensure high purity. Homogeneous iOPCs exhibit typical small-bipolar morphology, maintain their self-renewal capacity and OPC marker expression for more than 31 passages, share high similarity in the global gene expression profile to wild-type OPCs, and give rise to mature oligodendrocytes and astrocytes in vitro and in vivo. Notably, transplanted iOPCs contribute to functional recovery in a spinal cord injury (SCI) model without tumor formation. This study provides a simple strategy to generate functional self-renewing iOPCs and yields insights for the in-depth study of demyelination and regenerative medicine.


Subject(s)
Octamer Transcription Factor-3/metabolism , Oligodendroglia/metabolism , Oligodendroglia/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Stem Cells/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/cytology , Immunohistochemistry , Karyotype , Male , Mice , Mice, SCID , Octamer Transcription Factor-3/genetics , Oligodendroglia/cytology , Rats , Recovery of Function/physiology , Spinal Cord Injuries/genetics , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/physiology
9.
Mol Neurodegener ; 10: 38, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26293123

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is an inexorable neurodegenerative disease that commonly occurs in the elderly. The cognitive impairment caused by AD is associated with abnormal accumulation of amyloid-ß (Aß) and hyperphosphorylated tau, which are accompanied by inflammation. Neural stem cells (NSCs) are self-renewing, multipotential cells that differentiate into distinct neural cells. When transplanted into a diseased brain, NSCs repair and replace injured tissues after migration toward and engraftment within lesions. We investigated the therapeutic effects in an AD mouse model of human NSCs (hNSCs) that derived from an aborted human fetal telencephalon at 13 weeks of gestation. Cells were transplanted into the cerebral lateral ventricles of neuron-specific enolase promoter-controlled APPsw-expressing (NSE/APPsw) transgenic mice at 13 months of age. RESULTS: Implanted cells extensively migrated and engrafted, and some differentiated into neuronal and glial cells, although most hNSCs remained immature. The hNSC transplantation improved spatial memory in these mice, which also showed decreased tau phosphorylation and Aß42 levels and attenuated microgliosis and astrogliosis. The hNSC transplantation reduced tau phosphorylation via Trk-dependent Akt/GSK3ß signaling, down-regulated Aß production through an Akt/GSK3ß signaling-mediated decrease in BACE1, and decreased expression of inflammatory mediators through deactivation of microglia that was mediated by cell-to-cell contact, secretion of anti-inflammatory factors generated from hNSCs, or both. The hNSC transplantation also facilitated synaptic plasticity and anti-apoptotic function via trophic supplies. Furthermore, the safety and feasibility of hNSC transplantation are supported. CONCLUSIONS: These findings demonstrate the hNSC transplantation modulates diverse AD pathologies and rescue impaired memory via multiple mechanisms in an AD model. Thus, our data provide tangible preclinical evidence that human NSC transplantation could be a safe and versatile approach for treating AD patients.


Subject(s)
Alzheimer Disease/therapy , Fetal Tissue Transplantation , Neural Stem Cells/transplantation , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Lineage , Cell Movement , Disease Models, Animal , Gestational Age , Gliosis/prevention & control , Graft Survival , Heterografts , Humans , Lateral Ventricles , Mice , Mice, Transgenic , Mutation, Missense , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/genetics , Phosphorylation , Point Mutation , Protein Processing, Post-Translational , Signal Transduction , Spatial Memory , Telencephalon/cytology , tau Proteins/metabolism
10.
Opt Express ; 17(4): 2481-6, 2009 Feb 16.
Article in English | MEDLINE | ID: mdl-19219151

ABSTRACT

We fabricated an elliptical hollow-core photonic bandgap fiber (EC-PBGF) by controlling lateral tension in the hollow core region during the fiber drawing process. The absolute value of group modal birefringence becomes relatively high near the bandgap boundaries. We also experimentally measured the strain and temperature sensitivities of the fabricated EC-PBGF-based Sagnac loop interferometer. The strain and temperature sensitivities were very much dependent upon the wavelength. Moreover this PBGF-based interferometer can be a good sensor of physical parameters such as strain and temperature.


Subject(s)
Interferometry/instrumentation , Manometry/instrumentation , Optical Fibers , Thermography/instrumentation , Computer-Aided Design , Elastic Modulus , Equipment Design , Equipment Failure Analysis , Interferometry/methods , Manometry/methods , Reproducibility of Results , Sensitivity and Specificity , Stress, Mechanical , Thermography/methods
11.
Opt Express ; 17(3): 1268-73, 2009 Feb 02.
Article in English | MEDLINE | ID: mdl-19188954

ABSTRACT

We describe the fabrication of elliptical hollow-core photonic bandgap fibers (EC-PBGFs). It was shown that the aspect ratio of the hollow core can be controlled by tuning the negative pressure in the space between the intermediate preform cane and outer jacketing tube, and by placing this preform assembly off-center in the furnace, resulting in lateral tension during the final draw. Modal birefringences of fabricated PBGFs with different aspect ratio were measured using a Sagnac loop interferometer. For the elliptical hollow core PBGF with aspect ratio of 2.34, the modal birefringence was measured to be about 4.6x10(-2) at 1,550 nm.

SELECTION OF CITATIONS
SEARCH DETAIL
...