Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gene Ther ; 26(5): 135-150, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30692604

RESUMEN

Gene therapy technologies are inevitably required to boost the therapeutic performance of cell therapies; thus, validating the efficacy of gene carriers specifically used for preparing cellular therapeutics is a prerequisite for evaluating the therapeutic capabilities of gene and cell combinatorial therapies. Herein, the efficacy of a recombinant adeno-associated virus derivative (rAAVr3.45) was examined to evaluate its potential as a gene carrier for genetically manipulating interleukin-10 (IL10)-secreting human neural stem cells (hNSCs) that can potentially treat ischemic injuries or neurological disorders. Safety issues that could arise during the virus preparation or viral infection were investigated; no replication-competent AAVs were detected in the final cell suspensions, transgene expression was mostly transient, and no severe interference on endogenous gene expression by viral infection occurred. IL10 secretion from hNSCs infected by rAAVr3.45 encoding IL10 did not alter the transcriptional profile of any gene by more than threefold, but the exogenously boosted IL10 was sufficient to provoke immunomodulatory effects in an ischemic brain injury animal model, thereby accelerating the recovery of neurological deficits and the reduction of brain infarction volume. This study presents evidence that rAAVr3.45 can be potentially used as a gene carrier to prepare stem cell therapeutics.


Asunto(s)
Isquemia Encefálica/terapia , Dependovirus/genética , Terapia Genética/métodos , Interleucina-10/genética , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Terapia Genética/efectos adversos , Células HEK293 , Humanos , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos ICR , Células-Madre Neurales/metabolismo , Trasplante de Células Madre/efectos adversos
2.
Nano Lett ; 18(2): 838-845, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29393650

RESUMEN

Cell-based therapies are attractive for treating various degenerative disorders and cancer but delivering functional cells to the region of interest in vivo remains difficult. The problem is exacerbated in dense biological matrices such as solid tissues because these environments impose significant steric hindrances for cell movement. Here, we show that neural stem cells transfected with zinc-doped ferrite magnetic nanoparticles (ZnMNPs) can be pulled by an external magnet to migrate to the desired location in the brain. These magnetically labeled cells (Mag-Cells) can migrate because ZnMNPs generate sufficiently strong mechanical forces to overcome steric hindrances in the brain tissues. Once at the site of lesion, Mag-Cells show enhanced neuronal differentiation and greater secretion of neurotrophic factors than unlabeled control stem cells. Our study shows that ZnMNPs activate zinc-mediated Wnt signaling to facilitate neuronal differentiation. When implemented in a rodent brain stroke model, Mag-Cells led to significant recovery of locomotor performance in the impaired limbs of the animals. Our findings provide a simple magnetic method for controlling migration of stem cells with high therapeutic functions, offering a valuable tool for other cell-based therapies.


Asunto(s)
Encéfalo/citología , Diferenciación Celular , Movimiento Celular , Magnetismo/métodos , Nanopartículas de Magnetita/química , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Animales , Encéfalo/patología , Infarto Encefálico/patología , Infarto Encefálico/terapia , Rastreo Celular , Células Cultivadas , Compuestos Férricos/química , Humanos , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/terapia , Ratas , Zinc/química
3.
Neural Plast ; 2015: 630932, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26568892

RESUMEN

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.


Asunto(s)
Médula Cervical/lesiones , Células Madre Fetales/trasplante , Células-Madre Neurales/trasplante , Traumatismos de la Médula Espinal/rehabilitación , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/efectos adversos , Adolescente , Adulto , Femenino , Humanos , Extremidad Inferior/inervación , Extremidad Inferior/fisiopatología , Masculino , Persona de Mediana Edad , Actividad Motora , Espasticidad Muscular , Conducción Nerviosa , Dimensión del Dolor , Traumatismos de la Médula Espinal/patología , Resultado del Tratamiento , Extremidad Superior/inervación , Extremidad Superior/fisiopatología , Adulto Joven
4.
Materials (Basel) ; 14(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34576423

RESUMEN

In this study, the effect of internal pores formed by a superabsorbent polymer (SAP) was analyzed by evaluating the compressive strength, chloride penetration depth, drying shrinkage, and pore size distribution of SAP-containing concrete, while securing workability using a water-reducing agent (WRA). The experimental results showed that the amount of WRA necessary increased as the amount of SAP added increased, and that the compressive strength was the highest when the SAP content was 1.5% of the concrete mix. Drying shrinkage tended to decrease as the SAP content increased, and it decreased by approximately 31-41% when the SAP content was 2.0% compared to that of the reference mix. The SAP expanded by approximately three times inside concrete, and it was distributed within the internal pores of air-entrained concrete. The optimal SAP content in concrete mix was 1.5%, and an SAP content of 2.0% or higher adversely affected the workability and compressive strength.

5.
Materials (Basel) ; 13(22)2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33187220

RESUMEN

This study analyzed the fundamental properties of concrete using steel slag, to test its viability as an aggregate material. An experimental investigation into the effect of steel slag as a coarse aggregate, and heavyweight waste glass as a fine aggregate, on the drying shrinkage of concrete was performed. The calculated shrinkage strain was compared to five different shrinkage prediction models, namely, the ACI 209, B3, KCI 2012, EC 2 and GL 2000 model codes, to evaluate their ability to accurately predict shrinkage behavior. From the results, the elastic modulus of concrete increased with the increase in steel slag substitution ratio, however drying shrinkage decreased. The predictive value of the existing prediction model of drying shrinkage differed from the experimental values, and requires correction to improve its accuracy. The B3 model code showed the best prediction results of drying shrinkage.

6.
Cells ; 9(5)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403417

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/terapia , Hipoxia-Isquemia Encefálica/terapia , Células-Madre Neurales/trasplante , Factor de Necrosis Tumoral alfa/farmacología , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Conducta Animal/efectos de los fármacos , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Caspasa 3/metabolismo , Línea Celular , Polaridad Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CX3CL1/metabolismo , Medios de Cultivo Condicionados/farmacología , Ácido Glutámico/toxicidad , Humanos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/patología , Ratones Endogámicos ICR , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuroprotección/efectos de los fármacos , Fenotipo , Estrés Fisiológico/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
7.
Yonsei Med J ; 61(6): 492-505, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32469173

RESUMEN

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.


Asunto(s)
Hipoxia-Isquemia Encefálica/terapia , Ventrículos Laterales/citología , Células-Madre Neurales/citología , Neurogénesis , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Hipoxia-Isquemia Encefálica/patología , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/metabolismo , Nestina/metabolismo
8.
Exp Mol Med ; 41(7): 487-500, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19322031

RESUMEN

Neural progenitor cells (NPs) have shown several promising benefits for the treatment of neurological disorders. To evaluate the therapeutic potential of human neural progenitor cells (hNPs) in amyotrophic lateral sclerosis (ALS), we transplanted hNPs or growth factor (GF)-expressing hNPs into the central nervous system (CNS) of mutant Cu/Zn superoxide dismutase (SOD1(G93A)) transgenic mice. The hNPs were engineered to express brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1), VEGF, neurotrophin-3 (NT-3), or glial cell-derived neurotrophic factor (GDNF), respectively, by adenoviral vector and GDNF by lentiviral vector before transplantation. Donor-derived cells engrafted and migrated into the spinal cord or brain of ALS mice and differentiated into neurons, oligodendrocytes, or glutamate transporter-1 (GLT1)-expressing astrocytes while some cells retained immature markers. Transplantation of GDNF- or IGF-1-expressing hNPs attenuated the loss of motor neurons and induced trophic changes in motor neurons of the spinal cord. However, improvement in motor performance and extension of lifespan were not observed in all hNP transplantation groups compared to vehicle-injected controls. Moreover, the lifespan of GDNF-expressing hNP recipient mice by lentiviral vector was shortened compared to controls, which was largely due to the decreased survival times of female animals. These results imply that although implanted hNPs differentiate into GLT1-expressing astrocytes and secrete GFs, which maintain dying motor neurons, inadequate trophic support could be harmful and there is sexual dimorphism in response to GDNF delivery in ALS mice. Therefore, additional therapeutic approaches may be required for full functional recovery.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Encéfalo/embriología , Células Madre Fetales/metabolismo , Neuronas Motoras/fisiología , Factores de Crecimiento Nervioso/metabolismo , Trasplante de Células Madre , Adenoviridae/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Astrocitos/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/metabolismo , Femenino , Vectores Genéticos , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Transgénicos , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Transfección , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Exp Neurobiol ; 28(6): 679-696, 2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-31902156

RESUMEN

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.

10.
Exp Mol Med ; 50(4): 1-14, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29622770

RESUMEN

Neural progenitor cell (NPC) transplantation has been shown to be beneficial in the ischemic brain. However, the low survival rate of transplanted NPCs in an ischemic microenvironment limits their therapeutic effects. Tumor necrosis factor-alpha (TNF-α) is one of the proinflammatory cytokines involved in the pathogenesis of various injuries. On the other hand, several studies have shown that TNF-α influences the proliferation, survival, and differentiation of NPCs. Our study investigated the effect of TNF-α pretreatment on human NPCs (hNPCs) under ischemia-related conditions in vitro. hNPCs harvested from fetal brain tissue were pretreated with TNF-α before being subjected to oxygen-glucose deprivation (OGD) to mimic ischemia in vitro. TNF-α pretreatment improved the viability and reduced the apoptosis of hNPCs after OGD. At the molecular level, TNF-α markedly increased the level of NF-κB signaling in hNPCs, and an NF-κB pathway inhibitor, BAY11-7082, completely reversed the protective effects of TNF-α on hNPCs. These results suggest that TNF-α improves hNPC survival by activating the NF-κB pathway. In addition, TNF-α significantly enhanced the expression of cellular inhibitor of apoptosis 2 (cIAP2). Use of a lentivirus-mediated short hairpin RNA targeting cIAP2 mRNA demonstrated that cIAP2 protected against OGD-induced cytotoxicity in hNPCs. Our study of intracellular NF-κB signaling revealed that inhibition of NF-κB activity abolished the TNF-α-mediated upregulation of cIAP2 in hNPCs and blocked TNF-α-induced cytoprotection against OGD. Therefore, this study suggests that TNF-α pretreatment, which protects hNPCs from OGD-induced apoptosis by activating the NF-κB pathway, provides a safe and simple approach to improve the viability of transplanted hNPCs in cerebral ischemia.


Asunto(s)
Glucosa/metabolismo , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
11.
Exp Mol Med ; 50(4): 1-18, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29674624

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/terapia , Células Inmovilizadas/trasplante , Células-Madre Neurales/trasplante , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre , Andamios del Tejido/química , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Xenoinjertos , Humanos , Ratones , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
12.
Macromol Biosci ; 17(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28809089

RESUMEN

Devising efficient gene delivery systems is crucial to enhancing the therapeutic efficacy of gene-cell therapy approaches. Herein, inverted quasi-spherical (iQS) droplet systems, which enhance gene delivery efficiencies by reducing the path lengths of gene vectors, mediating motions of vectors at early stages, and raising the contact frequencies of vectors with cells, are developed by adopting the principle of 3D hanging-drop cell culture. Micrometer-sized polydopamine (pDA) holes are created on superhydrophobic titanium isopropoxide (TiO2 )-coated substrates by physical scraping; droplets are loaded on the pDA holes, and inversion of the substrate generates iQS droplets with large contact angles. Both human neural stem cells (hNSCs) and adeno-associated viral vectors are simultaneously incorporated into the iQS droplets to assess gene delivery efficiencies. The steep angles of iQS droplets and enhanced cell/vector contact frequencies facilitate the viral association with hNSCs and enhancing cell-cell interactions, thereby significantly promoting gene delivery efficiencies. Even with reduced viral quantities/exposure times and cell numbers, the iQS droplet systems elicit sufficient gene expression (i.e., interleukin-10). The ability of the iQS droplet systems to maximize beneficial gene delivery effects with minimal materials (e.g., medium, cells, and vectors) should enable their extensive use as a platform for preparing genetically stimulated cellular therapeutics.


Asunto(s)
Técnicas de Transferencia de Gen , Indoles/química , Polímeros/química , Titanio/química , Comunicación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Dependovirus , Vectores Genéticos/metabolismo , Humanos , Interleucina-10/farmacología , Células-Madre Neurales/metabolismo
13.
Transl Res ; 183: 121-136.e9, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28081931

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Lesiones Encefálicas/terapia , Hipoxia-Isquemia Encefálica/terapia , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Línea Celular Tumoral , Trasplante de Células , Regulación de la Expresión Génica , Humanos , Ratones , Células Madre Multipotentes , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Técnicas de Cultivo de Tejidos
14.
Mol Neurodegener ; 10: 38, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26293123

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/terapia , Trasplante de Tejido Fetal , Células-Madre Neurales/trasplante , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Linaje de la Célula , Movimiento Celular , Modelos Animales de Enfermedad , Edad Gestacional , Gliosis/prevención & control , Supervivencia de Injerto , Xenoinjertos , Humanos , Ventrículos Laterales , Ratones , Ratones Transgénicos , Mutación Missense , Fragmentos de Péptidos/metabolismo , Fosfopiruvato Hidratasa/genética , Fosforilación , Mutación Puntual , Procesamiento Proteico-Postraduccional , Transducción de Señal , Memoria Espacial , Telencéfalo/citología , Proteínas tau/metabolismo
15.
Macromol Biosci ; 15(10): 1348-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26036788

RESUMEN

Biophysical cues provided by nanotopographical surfaces have been used as stimuli to guide neurite extension and regulate neural stem cell (NSC) differentiation. Here, we fabricated biodegradable polymer substrates with nanoscale topography for enhancing human NSC (hNSC) differentiation and guided neurite outgrowth. The substrate was constructed from biodegradable poly(lactic-co-glycolic acid) (PLGA) using solvent-assisted capillary force lithography. We found that precoating with 3,4-dihydroxy-l-phenylalanine (DOPA) facilitated the immobilization of poly-l-lysine and fibronectin on PLGA substrates via bio-inspired catechol chemistry. The DOPA-coated nanopatterned substrates directed cellular alignment along the patterned grooves by contact guidance, leading to enhanced focal adhesion, skeletal protein reorganization, and neuronal differentiation of hNSCs as indicated by highly extended neurites from cell bodies and increased expression of neuronal markers (Tuj1 and MAP2). The addition of nerve growth factor further enhanced neuronal differentiation of hNSCs, indicating a synergistic effect of biophysical and biochemical cues on NSC differentiation. These bio-inspired PLGA nanopatterned substrates could potentially be used as implantable biomaterials for improving the efficacy of hNSCs in treating neurodegenerative diseases.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Nanopartículas/química , Factores de Crecimiento Nervioso/farmacología , Células-Madre Neurales/citología , Neuronas/citología , Biomarcadores/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ácido Láctico/química , Células-Madre Neurales/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuronas/efectos de los fármacos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Propiedades de Superficie , Agua/química
16.
Yonsei Med J ; 45 Suppl: 32-40, 2004 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15250048

RESUMEN

Neural stem cells (NSCs) are operationally defined by their ability to self-renew, to differentiate into cells of all glial and neuronal lineages throughout the neuraxis, and to populate developing or degenerating central nervous system (CNS) regions. Thus their use as graft material can be considered analogous to hematopoietic stem cell-mediated reconstitution and gene transfer. The recognition that NSCs propagated in culture could be reimplanted into mammalian brain, where they might integrate appropriately throughout the mammalian CNS and stably express foreign genes, has unveiled a new role for neural transplantation and gene therapy and a possible strategy for addressing the CNS manifestations of diseases that heretofore had been refractory to intervention. NSCs additionally have the appealing ability to home in on pathology, even over great distances. Such observations help to advance the idea that NSCs--as a prototype for stem cells from other solid organs--might aid in reconstructing the molecular and cellular milieu of maldeveloped or damaged CNS.


Asunto(s)
Terapia Genética/métodos , Tejido Nervioso/citología , Trasplante de Células Madre , Células Madre/metabolismo , Animales , Técnicas de Transferencia de Gen , Humanos , Células Madre/fisiología
17.
Sci Rep ; 4: 6319, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-25204726

RESUMEN

Neural stem cells (NSCs) are characterized by a capacity for self-renewal, differentiation into multiple neural lineages, all of which are considered to be promising components for neural regeneration. However, for cell-replacement therapies, it is essential to monitor the process of in vitro NSC differentiation and identify differentiated cell phenotypes. We report a real-time and label-free method that uses a capacitance sensor array to monitor the differentiation of human fetal brain-derived NSCs (hNSCs) and to identify the fates of differentiated cells. When hNSCs were placed under proliferation or differentiation conditions in five media, proliferating and differentiating hNSCs exhibited different frequency and time dependences of capacitance, indicating that the proliferation and differentiation status of hNSCs may be discriminated in real-time using our capacitance sensor. In addition, comparison between real-time capacitance and time-lapse optical images revealed that neuronal and astroglial differentiation of hNSCs may be identified in real-time without cell labeling.


Asunto(s)
Astrocitos/citología , Encéfalo/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Proliferación Celular/fisiología , Células Cultivadas , Capacidad Eléctrica , Estimulación Eléctrica , Humanos
18.
PLoS One ; 9(8): e104092, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25105891

RESUMEN

Cell transplantation has been suggested as an alternative therapy for temporal lobe epilepsy (TLE) because this can suppress spontaneous recurrent seizures in animal models. To evaluate the therapeutic potential of human neural stem/progenitor cells (huNSPCs) for treating TLE, we transplanted huNSPCs, derived from an aborted fetal telencephalon at 13 weeks of gestation and expanded in culture as neurospheres over a long time period, into the epileptic hippocampus of fully kindled and pilocarpine-treated adult rats exhibiting TLE. In vitro, huNSPCs not only produced all three central nervous system neural cell types, but also differentiated into ganglionic eminences-derived γ-aminobutyric acid (GABA)-ergic interneurons and released GABA in response to the depolarization induced by a high K+ medium. NSPC grafting reduced behavioral seizure duration, afterdischarge duration on electroencephalograms, and seizure stage in the kindling model, as well as the frequency and the duration of spontaneous recurrent motor seizures in pilocarpine-induced animals. However, NSPC grafting neither improved spatial learning or memory function in pilocarpine-treated animals. Following transplantation, grafted cells showed extensive migration around the injection site, robust engraftment, and long-term survival, along with differentiation into ß-tubulin III+ neurons (∼34%), APC-CC1+ oligodendrocytes (∼28%), and GFAP+ astrocytes (∼8%). Furthermore, among donor-derived cells, ∼24% produced GABA. Additionally, to explain the effect of seizure suppression after NSPC grafting, we examined the anticonvulsant glial cell-derived neurotrophic factor (GDNF) levels in host hippocampal astrocytes and mossy fiber sprouting into the supragranular layer of the dentate gyrus in the epileptic brain. Grafted cells restored the expression of GDNF in host astrocytes but did not reverse the mossy fiber sprouting, eliminating the latter as potential mechanism. These results suggest that human fetal brain-derived NSPCs possess some therapeutic effect for TLE treatments although further studies to both increase the yield of NSPC grafts-derived functionally integrated GABAergic neurons and improve cognitive deficits are still needed.


Asunto(s)
Encéfalo/metabolismo , Diferenciación Celular/fisiología , Epilepsia del Lóbulo Temporal/terapia , Feto/citología , Células-Madre Neurales/trasplante , Análisis de Varianza , Animales , Western Blotting , Encéfalo/citología , Cromatografía Líquida de Alta Presión , Electroencefalografía , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Fibras Musgosas del Hipocampo/metabolismo , Pilocarpina , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Resultado del Tratamiento , Ácido gamma-Aminobutírico/metabolismo
19.
ACS Nano ; 8(4): 3393-401, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24673493

RESUMEN

One of the holy grails in biomedical imaging technology is to achieve accurate imaging of biological targets. The development of sophisticated instrumentation and the use of contrast agents have improved the accuracy of biomedical imaging. However, the issue of false imaging remains a problem. Here, we developed a dual-mode artifact filtering nanoparticle imaging agent (AFIA) that comprises a combination of paramagnetic and superparamagnetic nanomaterials. This AFIA has the ability to perform "AND logic gate" algorithm to eliminate false errors (artifacts) from the raw images to enhance accuracy of the MRI. We confirm the artifact filtering capability of AFIA in MRI phantoms and further demonstrate that artifact-free imaging of stem cell migration is possible in vivo.


Asunto(s)
Artefactos , Medios de Contraste , Ingeniería , Imagen por Resonancia Magnética/métodos , Nanopartículas , Animales , Encéfalo/citología , Rastreo Celular , Medios de Contraste/química , Células Madre Embrionarias/citología , Femenino , Humanos , Imanes/química , Manganeso/química , Modelos Moleculares , Conformación Molecular , Nanopartículas/química , Compuestos Organometálicos/química , Embarazo , Ratas
20.
Exp Mol Med ; 45: e60, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24232259

RESUMEN

Alzheimer's disease (AD) is the most common cause of age-related dementia. The neuropathological hallmarks of AD include extracellular deposition of amyloid-ß peptides and neurofibrillary tangles that lead to intracellular hyperphosphorylated tau in the brain. Soluble amyloid-ß oligomers are the primary pathogenic factor leading to cognitive impairment in AD. Neural stem cells (NSCs) are able to self-renew and give rise to multiple neural cell lineages in both developing and adult central nervous systems. To explore the relationship between AD-related pathology and the behaviors of NSCs that enable neuroregeneration, a number of studies have used animal and in vitro models to investigate the role of amyloid-ß on NSCs derived from various brain regions at different developmental stages. However, the Aß effects on NSCs remain poorly understood because of conflicting results. To investigate the effects of amyloid-ß oligomers on human NSCs, we established amyloid precursor protein Swedish mutant-expressing cells and identified cell-derived amyloid-ß oligomers in the culture media. Human NSCs were isolated from an aborted fetal telencephalon at 13 weeks of gestation and expanded in culture as neurospheres. Human NSCs exposure to cell-derived amyloid-ß oligomers decreased dividing potential resulting from senescence through telomere attrition, impaired neurogenesis and promoted gliogenesis, and attenuated mobility. These amyloid-ß oligomers modulated the proliferation, differentiation and migration patterns of human NSCs via a glycogen synthase kinase-3ß-mediated signaling pathway. These findings contribute to the development of human NSC-based therapy for AD by elucidating the effects of Aß oligomers on human NSCs.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Células-Madre Neurales/efectos de los fármacos , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Senescencia Celular , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Feto/citología , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Transducción de Señal , Acortamiento del Telómero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA