Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 45(6): 531-537, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30636067

RESUMO

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is an early-onset neurodegenerative disease that includes progressive cerebellar dysfunction. ARSACS is caused by an autosomal recessive loss-of-function mutation in the SACS gene, which encodes for SACSIN. Although animal models are still necessary to investigate the role of SACSIN in the pathology of this disease, more reliable human cellular models need to be generated to better understand the cerebellar pathophysiology of ARSACS. The discovery of human induced pluripotent stem cells (hiPSC) has permitted the derivation of patient-specific cells. These cells have an unlimited self-renewing capacity and the ability to differentiate into different neural cell types, allowing studies of disease mechanism, drug discovery and cell replacement therapies. In this study, we discuss how the hiPSC-derived cerebellar organoid culture offers novel strategies for targeting the pathogenic mutations related to ARSACS. We also highlight the advantages and challenges of this 3D cellular model, as well as the questions that still remain unanswered.


Assuntos
Doenças Cerebelares/patologia , Cerebelo/patologia , Espasticidade Muscular/patologia , Ataxias Espinocerebelares/congênito , Animais , Doenças Cerebelares/terapia , Humanos , Células-Tronco Pluripotentes Induzidas , Modelos Teóricos , Espasticidade Muscular/terapia , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/terapia
2.
Bull Exp Biol Med ; 155(4): 536-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24143385

RESUMO

A central issue in stem cell biology is a better understanding of the molecular mechanisms that regulate self-renewal of human hematopoietic stem cells (HSCs). Control of the specific function of HSCs like self-renewal and differentiation might be regulated by a common set of critical genes. However, the regulation among these genes is yet to be elucidated. Here, we show that activation by a novel human GPI-linked glycoprotein ACA at the surface of human peripheral blood progenitor cells induces via PI3K/Akt/mTor/PTEN upregulation of WNT, Notch1, Bmi-1 and HoxB4 genes thus, promoting self-renewal and generation of primitive HSCs. ACA-generated self-renewing cells retained their lympho-myeloid repopulating potential in NOD/SCID mouse xeno-transplantation model with long term functional capacity. We conclude that ACA is an essential regulator of the genes involved in maintaining hematopoiesis and its use in clinical praxis could overcome many of the barriers present so far in transplantation medicine.


Assuntos
Proteínas Sanguíneas/fisiologia , Hematopoese , Glicoproteínas de Membrana/fisiologia , Animais , Antígenos CD34/metabolismo , Proliferação de Células , Células Cultivadas , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Xenoenxertos , Humanos , Leucócitos Mononucleares/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fosforilação , Processamento de Proteína Pós-Traducional , Regulação para Cima , Via de Sinalização Wnt
3.
Bull Exp Biol Med ; 155(4): 552-67, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24143386

RESUMO

Reprogramming of human somatic cells by transcription factors to pluripotent state holds great promise for regenerative medicine. However, low efficiencies of current reprogramming methods, immunogenicity and lack of understanding regarding the molecular mechanisms responsible for their generation, limits their utilization and raises questions regarding safety for therapeutic application. Here we report that ACA signaling via PI3K/Akt/mTor induces sustained de-differentiation of human blood progenitor cells leading to generation of ACA pluripotent stem cells. Blood-derived pluripotent stem cells differentiate in vitro into cell types of all three germ layers, exhibiting neuronal, liver, or endothelial characteristics. Our results reveal insight into the molecular events regulating cellular reprogramming and also indicate that pluripotency might be controlled in vivo through binding of a natural ligand(s) to ACA receptor enabling reprogramming through defined pathway(s) and providing a safe and efficient method for generation of pluripotent stem cells which could be a breakthrough in human therapeutics.


Assuntos
Proteínas Sanguíneas/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Glicoproteínas de Membrana/fisiologia , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Sangue Fetal/citologia , Humanos , Imunofenotipagem , Células-Tronco Pluripotentes Induzidas/transplante , Leucócitos Mononucleares/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neurônios/metabolismo , Oócitos/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...