Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Prolif ; 56(12): e13500, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37246473

RESUMO

The activation of hepatic stellate cells (HSCs) is the main cause of liver fibrogenesis in response to different etiologies of chronic liver injuries. HSCs are heterogeneous, but the lack of specific markers to distinguish different HSC subset hinders the development of targeted therapy for liver fibrosis. In this study, we aim to reveal new HSC subsets by cell fate tracking. We constructed a novel ReelinCreERT2 transgenic mouse model to track the fate of cells expressing Reelin and their progeny (Reelin+ cells). And we investigated the property of Reelin+ cells, such as differentiation and proliferation, in hepatotoxic (carbon tetrachloride; CCl4 ) or cholestatic (bile duct ligation; BDL) liver injury models by immunohistochemistry. Our study revealed that Reelin+ cells were a new HSC subset. In terms of activation, migration, and proliferation, Reelin+ HSCs displayed different properties from Desmin+ HSCs (total HSCs) in cholestatic liver injury model but shared similar properties to total HSCs in hepatotoxic liver injury model. Besides, we did not find evidence that Reelin+ HSCs transdifferentiated into hepatocytes or cholangiocytes through mesenchymal-epithelial transition (MET). In this study, our genetic cell fate tracking data reveal that ReelinCreERT2-labelled cells are a new HSC subset, which provides new insights into targeted therapy for liver fibrosis.


Assuntos
Células Estreladas do Fígado , Fígado , Camundongos , Animais , Células Estreladas do Fígado/patologia , Desmina , Fígado/patologia , Cirrose Hepática/patologia , Camundongos Transgênicos , Tetracloreto de Carbono/toxicidade , Proliferação de Células
2.
Stem Cell Reports ; 15(3): 706-720, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32763157

RESUMO

Liver progenitor cells (LPCs) have a remarkable contribution to the hepatocytes and ductal cells when normal hepatocyte proliferation is severely impaired. As a biomarker for LPCs, Sry-box 9 (Sox9) plays critical roles in liver homeostasis and repair in response to injury. However, the regulation mechanism of Sox9 in liver physiological and pathological state remains unknown. In this study, we found that miR-126 positively regulated the expression of Sox9, the proliferation and differentiation of SOX9+ LPCs by suppressing the translation of homeobox b6 (Hoxb6). As a transcription factor, HOXB6 directly binds to the promoter of Sox9 to inhibit Sox9 expression, resulting in the destruction of the properties of SOX9+ LPCs in CCl4-induced liver injury. These findings revealed the role of miR-126 in regulating SOX9+ LPCs fate by targeting Hoxb6 in liver injury repair. Our findings suggest the potential role of miR-126 as a nucleic acid therapy drug target for liver failure.


Assuntos
Proteínas de Homeodomínio/metabolismo , Regeneração Hepática , Fígado/metabolismo , MicroRNAs/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/metabolismo , Animais , Sequência de Bases , Tetracloreto de Carbono , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Doença Crônica , Dependovirus/metabolismo , Modelos Animais de Doenças , Hepatócitos/citologia , Fígado/lesões , Fígado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3
3.
Biotechnol Lett ; 42(8): 1327-1336, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32221722

RESUMO

OBJECTIVES: Recent research has indicated the microRNA-126a (miR-126a) is an endothelial cell-specific and highly conserved endogenous small non-coding RNA molecule. It contributes to the vascular integrity and angiogenesis, but the molecular regulation mechanism of miR-126a remains unknown. RESULTS: Herein, quantitative real-time polymerase chain reaction (qRT-PCR) results showed that Farnesoid X Receptor (FXR) activation promoted miR-126a expression in HepG2, LO2, and Hep1-6 cells. Furthermore, FXR was found to transcriptionally regulate the miR-126a by binding to its DR8 site. The binding site of FXR was confirmed on intron 6 or 7 of miR-126a host gene epidermal growth factor-like domain 7 (EGFL7) by luciferase reporter assays, electrophoretic mobility shift assays (EMSAs) and chromatin immunoprecipitation (ChIP) assays. CONCLUSIONS: All these data collectively suggest that FXR regulates transcripts of miR-126a by binding to DR8 in miR-126a gene promoter. This study may provide a molecular therapeutic target for angiogenic disorders, aging, and liver failure.


Assuntos
Regulação da Expressão Gênica/genética , MicroRNAs , Receptores Citoplasmáticos e Nucleares , Animais , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular , Família de Proteínas EGF/genética , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcrição Gênica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...