Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Yonsei Medical Journal ; : 492-505, 2020.
Artigo | WPRIM | ID: wpr-833360

RESUMO

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.

2.
Experimental Neurobiology ; : 679-696, 2019.
Artigo em Inglês | WPRIM | ID: wpr-785789

RESUMO

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 line-derived 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 GDNF-hNSPCs 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.


Assuntos
Animais , Humanos , Ratos , Axônios , Morte Celular , Movimento Celular , Terapia Baseada em Transplante de Células e Tecidos , Cicatriz , Doenças Desmielinizantes , Ácido gama-Aminobutírico , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hiperalgesia , Bainha de Mielina , Neuralgia , Neuritos , Neuroglia , Neurônios , Neuropeptídeo Y , Paraplegia , Tratos Piramidais , Regeneração , Traumatismos da Medula Espinal , Medula Espinal , Usos Terapêuticos , Transplantes , Canais de Sódio Disparados por Voltagem
3.
Experimental & Molecular Medicine ; : e60-2013.
Artigo em Inglês | WPRIM | ID: wpr-152455

RESUMO

Alzheimer's disease (AD) is the most common cause of age-related dementia. The neuropathological hallmarks of AD include extracellular deposition of amyloid-beta peptides and neurofibrillary tangles that lead to intracellular hyperphosphorylated tau in the brain. Soluble amyloid-beta 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-beta on NSCs derived from various brain regions at different developmental stages. However, the Abeta effects on NSCs remain poorly understood because of conflicting results. To investigate the effects of amyloid-beta oligomers on human NSCs, we established amyloid precursor protein Swedish mutant-expressing cells and identified cell-derived amyloid-beta 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-beta oligomers decreased dividing potential resulting from senescence through telomere attrition, impaired neurogenesis and promoted gliogenesis, and attenuated mobility. These amyloid-beta oligomers modulated the proliferation, differentiation and migration patterns of human NSCs via a glycogen synthase kinase-3beta-mediated signaling pathway. These findings contribute to the development of human NSC-based therapy for AD by elucidating the effects of Abeta oligomers on human NSCs.


Assuntos
Animais , Humanos , Camundongos , Peptídeos beta-Amiloides/farmacologia , Apoptose , Senescência Celular , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados/química , Feto/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Células HEK293 , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Transdução de Sinais , Encurtamento do Telômero
4.
Neonatal Medicine ; : 343-353, 2013.
Artigo em Coreano | WPRIM | ID: wpr-97611

RESUMO

Neural stem cells (NSCs) are characterized by a capacity for self-renewal, differentiation into multiple neural cell lineages, and migration toward damaged sites in the central nervous system (CNS). NSCs expanded in culture could be implanted into the brain where they integrate into host neural circuitry and stably express foreign genes. It hence appears that transplantation of NSCs has been proposed as a promising therapeutic strategy in neurological disorders. During hypoxic-ischemic (HI) brain injury, factors are transiently elaborated to which NSCs respond by migrating to degenerating regions and differentiating towards replacement of dying neural cells. In addition, NSCs serve as vehicles for gene delivery and appear capable of simultaneous neural cell replacement and gene therapy (e.g. with factors that might enhance neuronal differentiation, neurites outgrowth, proper connectivity, neuroprotection, and/or immunomodulatory substances). When combined with certain synthetic biomaterials, NSCs may be even more effective in 'engineering' the damaged CNS towards reconstitution. Human NSCs were isolated from the forebrain of an aborted fetus at 13 weeks of gestation and were grown as neurospheres in cultures. After the characterization of human NSCs in preclinical testing and the approval of the IRB, a clinical trial of the transplantation of human NSCs into patients with severe perinatal HI brain injury has been performed. The existing data from these clinical trials have shown to be safe, well tolerated, and of neurologically-some benefits. Therefore, long-term and large scale multicenter clinical study is required to determine its precise therapeutic effect and safety.


Assuntos
Humanos , Gravidez , Feto Abortado , Materiais Biocompatíveis , Encéfalo , Lesões Encefálicas , Linhagem da Célula , Sistema Nervoso Central , Comitês de Ética em Pesquisa , Terapia Genética , Doenças do Sistema Nervoso , Células-Tronco Neurais , Neuritos , Neurônios , Prosencéfalo , Terapia Baseada em Transplante de Células e Tecidos , Transplantes
5.
Experimental & Molecular Medicine ; : 487-500, 2009.
Artigo em Inglês | WPRIM | ID: wpr-107287

RESUMO

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 (SOD(1G93A)) 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.


Assuntos
Animais , Feminino , Humanos , Masculino , Camundongos , Adenoviridae/genética , Esclerose Lateral Amiotrófica/metabolismo , Astrócitos/metabolismo , Encéfalo/embriologia , Diferenciação Celular , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/metabolismo , Células-Tronco Fetais/metabolismo , Vetores Genéticos , Técnicas Imunoenzimáticas , Camundongos Transgênicos , Neurônios Motores/fisiologia , Fatores de Crescimento Neural/metabolismo , Transplante de Células-Tronco , Superóxido Dismutase/genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética
6.
Yonsei Medical Journal ; : S32-S40, 2004.
Artigo em Inglês | WPRIM | ID: wpr-177679

RESUMO

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.


Assuntos
Animais , Humanos , Terapia Genética/métodos , Técnicas de Transferência de Genes , Tecido Nervoso/citologia , Transplante de Células-Tronco , Células-Tronco/metabolismo
7.
Journal of the Korean Radiological Society ; : 239-243, 1993.
Artigo em Coreano | WPRIM | ID: wpr-88753

RESUMO

Adult intussusception is a rare condition, unlike in children. Ihere is an identifiable bowel lesion as a leading point in most of cases. Retrospectively we reviewed 22 patients with adult intussusception, and analyzed CT scans of 13 patients during the last 5 years. Twenty of the 22 patients had various causes of intussusception and 7 patients were associated with primary malignancy, 5 patients with benign tumor, and 4 patients with adhesive band. CT scans of 13 cases all showed characteristic target appearance or sausage-like soft tissue mass within the bowel, suggesting intussusception. In eleven of the 13 patients(84.6%) an intussuscepting mass was identified on CT scan as the lead point. Specific diagnosis for the cause of intussusception could be made by CT in four of 13 cases (three lipomas and one mucocele). In conclusion, CT is useful not only for the diagnosis but also for the evaluation of the leading causes of intussusception. Especially, CT could determined the exact cause of intussusception in cases of lipoma and mucocele.


Assuntos
Adulto , Criança , Humanos , Adesivos , Diagnóstico , Intussuscepção , Lipoma , Mucocele , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA