Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Journal of Korean Medical Science ; : 171-177, 2016.
Artigo em Inglês | WPRIM | ID: wpr-133743

RESUMO

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson's disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3-4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6-8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10-15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson's disease.


Assuntos
Animais , Humanos , Camundongos , Encéfalo/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/química , Polpa Dentária/citologia , Neurônios Dopaminérgicos/citologia , Ensaio de Imunoadsorção Enzimática , Proteína Glial Fibrilar Ácida/genética , Camundongos Endogâmicos ICR , Proteína Básica da Mielina/genética , Reação em Cadeia da Polimerase em Tempo Real , Antígenos Embrionários Estágio-Específicos/genética , Células-Tronco/citologia , Tubulina (Proteína)/genética , Tirosina 3-Mono-Oxigenase/análise
2.
Journal of Korean Medical Science ; : 171-177, 2016.
Artigo em Inglês | WPRIM | ID: wpr-133742

RESUMO

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson's disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3-4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6-8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10-15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson's disease.


Assuntos
Animais , Humanos , Camundongos , Encéfalo/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/química , Polpa Dentária/citologia , Neurônios Dopaminérgicos/citologia , Ensaio de Imunoadsorção Enzimática , Proteína Glial Fibrilar Ácida/genética , Camundongos Endogâmicos ICR , Proteína Básica da Mielina/genética , Reação em Cadeia da Polimerase em Tempo Real , Antígenos Embrionários Estágio-Específicos/genética , Células-Tronco/citologia , Tubulina (Proteína)/genética , Tirosina 3-Mono-Oxigenase/análise
3.
Iranian Journal of Pediatrics. 2013; 23 (4): 481-484
em Inglês | IMEMR | ID: emr-138357

RESUMO

Alexander disease [AD] is a sporadic leukodystrophy that predominantly affects infants and children and usually results in death within ten years after onset. The infantile form comprises the most of affected individuals. It presents in the first two years of life, typically with progressive psychomotor retardation with loss of developmental milestones, megalencephaly and frontal bossing, seizures, pyramidal signs and ataxia. The diagnosis is based on magnetic resonance imaging [MRI] findings and confirmed by GFAP gene molecular testing. GFAP gene encodes glial fibrillary acidic protein, is the only gene in which mutation is currently known to cause AD which is inherited in autosomal dominant manner. In this article we report the first two Iranian cases of infantile AD and their clinical, brain MRI and molecular findings. We report two novel mutations too in the GFAP gene that are associated with infantile form of AD. GFAP gene mutations are a reliable marker for infantile AD diagnosed according to clinical and MRI defined criteria. A genotype-phenotype correlation had been discerned for the two most frequently reported GFAP gene mutations in infantile type of AD [R79 and R239], with the phenotype of the R79 mutations appearing much less severe than that of the R239 mutations. Our findings confirm this theory


Assuntos
Humanos , Masculino , Proteína Glial Fibrilar Ácida/genética , Estudos de Associação Genética , Leucoencefalopatias , Espectroscopia de Ressonância Magnética , Imageamento por Ressonância Magnética , Megalencefalia , Literatura de Revisão como Assunto
4.
Experimental & Molecular Medicine ; : e53-2013.
Artigo em Inglês | WPRIM | ID: wpr-209542

RESUMO

The transplantation of neural precursor cells (NPCs) is known to be a promising approach to ameliorating behavioral deficits after stroke in a rodent model of middle cerebral artery occlusion (MCAo). Previous studies have shown that transplanted NPCs migrate toward the infarct region, survive and differentiate into mature neurons to some extent. However, the spatiotemporal dynamics of NPC migration following transplantation into stroke animals have yet to be elucidated. In this study, we investigated the fates of human embryonic stem cell (hESC)-derived NPCs (ENStem-A) for 8 weeks following transplantation into the side contralateral to the infarct region using 7.0T animal magnetic resonance imaging (MRI). T2- and T2*-weighted MRI analyses indicated that the migrating cells were clearly detectable at the infarct boundary zone by 1 week, and the intensity of the MRI signals robustly increased within 4 weeks after transplantation. Afterwards, the signals were slightly increased or unchanged. At 8 weeks, we performed Prussian blue staining and immunohistochemical staining using human-specific markers, and found that high percentages of transplanted cells migrated to the infarct boundary. Most of these cells were CXCR4-positive. We also observed that the migrating cells expressed markers for various stages of neural differentiation, including Nestin, Tuj1, NeuN, TH, DARPP-32 and SV38, indicating that the transplanted cells may partially contribute to the reconstruction of the damaged neural tissues after stroke. Interestingly, we found that the extent of gliosis (glial fibrillary acidic protein-positive cells) and apoptosis (TUNEL-positive cells) were significantly decreased in the cell-transplanted group, suggesting that hESC-NPCs have a positive role in reducing glia scar formation and cell death after stroke. No tumors formed in our study. We also performed various behavioral tests, including rotarod, stepping and modified neurological severity score tests, and found that the transplanted animals exhibited significant improvements in sensorimotor functions during the 8 weeks after transplantation. Taken together, these results strongly suggest that hESC-NPCs have the capacity to migrate to the infarct region, form neural tissues efficiently and contribute to behavioral recovery in a rodent model of ischemic stroke.


Assuntos
Animais , Humanos , Masculino , Ratos , Apoptose , Diferenciação Celular , Movimento Celular , Células-Tronco Embrionárias/citologia , Proteína Glial Fibrilar Ácida/genética , Infarto da Artéria Cerebral Média/metabolismo , Células-Tronco Neurais/citologia , Desempenho Psicomotor , Ratos Sprague-Dawley , Receptores CXCR4/genética
6.
Journal of Veterinary Science ; : 1-6, 2011.
Artigo em Inglês | WPRIM | ID: wpr-224358

RESUMO

Intermediate filaments, including nestin and glial fibrillary acidic protein (GFAP), are important for the brain to accommodate neural activities and changes during development. The present study examined the temporal changes of nestin and GFAP protein levels in the postnatal development of the mouse hippocampus. Mouse hippocampi were sampled on postnatal day (PND) 1, 3, 6, 18, and 48. Western blot analysis showed that nestin expression was high at PND 1 and markedly decreased until PND 18. Conversely, GFAP expression was acutely increased in the early phase of postnatal development. Nestin immunoreactivity was localized mainly in the processes of ramified cells at PND 1, but expression subsequently decreased. In contrast, GFAP was evident mainly in the marginal cells of the hippocampus at PND 1, but immunoreactivity revealed satellite, radial, or ramified shapes of the cells from PND 6-48. This study demonstrates that the opposing pattern of nestin and GFAP expressions in mouse hippocampus during postnatal development occur in the early development stage (PND 1-18), suggesting that the opposing change of nestin and GFAP in early postnatal development is important for neural differentiation and positioning in the mouse hippocampus.


Assuntos
Animais , Feminino , Masculino , Camundongos , Envelhecimento , Western Blotting , Encéfalo/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteína Glial Fibrilar Ácida/genética , Hipocampo/citologia , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/genética , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo
7.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 21-4, 2006.
Artigo em Inglês | WPRIM | ID: wpr-634299

RESUMO

To examine the effects of ischemia and anoxia on cell activation and cell cycle of astrocytes in vitro, the cell cycles and the proliferation of astrocytes in different time points after ischemia and anoxia were studied by flow cytometry and BrdU labeling and the expression of GFAP and cyclin D1 was detected by the fluorescence immunochemistry. After ischemia and anoxia in vitro, the astrocytes in S phase were significantly increased as compared with those in the normal group and the proliferating ability of the astrocytes was highest 6 h after the treatment as revealed by BrdU pulse labeling, but the astrocytes in S phase and proliferating ability were decreased after 6 h. At the early stages of ischemia and anoxia, the positive staining intensity of GFAP was increased, peaked at 6th h, while 12 h after the ischemia and anoxia, the positive staining intensity of GFAP became weak, and the expression of cyclin D1 was gradually increased after the ischemic and anoxic damage. It is concluded that astrocytes are activated to proliferate and enter new cycle events by ischemia and anoxia, and cyclin D1 is implicated in the proliferation and repair of astrocytes. The cell cycle events are closely associated with the proliferation and activation of astrocytes.


Assuntos
Animais Recém-Nascidos , Astrócitos/citologia , Ciclo Celular , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Ciclina D1/biossíntese , Ciclina D1/genética , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Ratos Wistar
8.
Journal of Korean Medical Science ; : 954-957, 2006.
Artigo em Inglês | WPRIM | ID: wpr-195933

RESUMO

Alexander disease (AD) is a rare leukodystrophy of the central nervous system of unknown etiology. AD is characterized by progressive failure of central myelination and the accumulation of Rosenthal fibers in astrocytes, and is inevitably lethal in nature. Symptomatically, AD is associated with leukoencephalopathy with macrocephaly, seizures, and psychomotor retardation in infants, and usually leads to death within the first decade. Its characteristic magnetic resonance imaging (MRI) findings have been described as demyelination predominantly in the frontal lobe. Moreover, dominant mutations in the GFAP gene, coding for glial fibrillary acidic protein (GFAP), a principal astrocytic intermediate filament protein, have been shown to lead to AD. The disease can now be detected by genetic diagnosis. We report the Korean case of an 8-month-old male patient with AD. He was clinically characterized due to the presence of psychomotor retardation, megalencephaly, spasticity, and recurrent seizures including infantile spasms which is a remarkable presentation. Demyelination in the frontal lobe and in a portion of the temporal lobe was demonstrated by brain MRI. Moreover, DNA analysis of peripheral blood showed the presence of a R239L mutation in the GFAP gene, involving the replacement of guanine with thymine.


Assuntos
Masculino , Lactente , Humanos , Espasmos Infantis/etiologia , Mutação , Imageamento por Ressonância Magnética , Proteína Glial Fibrilar Ácida/genética , Eletroencefalografia , Doença de Alexander/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA