Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochem Soc Trans ; 51(3): 1271-1277, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37264940

RESUMO

The liver is a highly organized organ that consists of hepatic parenchymal cells, hepatocytes, and non-parenchymal cells such as the liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), cholangiocytes, and Kupffer cells. Although previous studies have primarily focused on the hepatocyte dynamics in the injured liver, recent studies have shown that non-parenchymal cells play an essential role in both liver regeneration and liver fibrosis progression. Among the non-parenchymal cells, HSCs directly contribute to the progression of liver fibrosis because the activation of HSCs in response to liver injury or inflammation results in the excess production of extra cellular matrix. LSECs also contribute to modulate the function of hepatocytes, HSCs, and immune cells during liver fibrosis. Therefore, to investigate the mechanisms for liver fibrosis in vitro, it is necessary to develop an appropriate liver model that accurately recapitulates the pathology of human liver fibrosis including HSC activation. However, the supply of human cells is limited and freshly isolated liver cells easily lose their specific characteristics in culture. To overcome this shortage of human liver cells, human induced pluripotent stem cell (hiPSC)-derived liver cells were generated by mimicking the liver developmental process. In this review article, we outline the differentiation system of liver non-parenchymal cells from hiPSCs and development of in vitro liver disease models using hiPSC-derived liver cells. We describe the utility of these liver models as experimental systems to investigate the mechanism of liver fibrosis and development of drugs for the treatment thereof.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células Endoteliais , Hepatócitos , Cirrose Hepática/terapia , Fígado
2.
Biol Open ; 11(9)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36017733

RESUMO

Recent genetic lineage tracing studies reveal heterogeneous origins of vascular endothelial cells and pericytes in the developing brain vasculature, despite classical experimental evidence for a mesodermal origin. Here we provide evidence through a genetic lineage tracing experiment that cephalic paraxial mesodermal cells give rise to endothelial cells and pericytes in the developing mouse brain. We show that Hepatic leukemia factor (Hlf) is transiently expressed by cephalic paraxial mesenchyme at embryonic day (E) 8.0-9.0 and the genetically marked E8.0 Hlf-expressing cells mainly contribute to the developing brain vasculature. Interestingly, the genetically marked E10.5 Hlf-expressing cells, which have been previously reported to contain embryonic hematopoietic stem cells, fail to contribute to the vascular cells. Combined, our genetic lineage tracing data demonstrate that a transient expression of Hlf marks a cephalic paraxial mesenchyme contributing to the developing brain vasculature. This article has an associated First Person interview with the first author of the paper.


Assuntos
Células Endoteliais , Leucemia , Animais , Encéfalo , Humanos , Leucemia/metabolismo , Mesoderma , Camundongos , Células-Tronco
3.
Stem Cell Reports ; 16(12): 3050-3063, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34861166

RESUMO

Hepatic stellate cells (HSCs) play a central role in the progression of liver fibrosis by producing extracellular matrices. The development of drugs to suppress liver fibrosis has been hampered by the lack of human quiescent HSCs (qHSCs) and an appropriate in vitro model that faithfully recapitulates HSC activation. In the present study, we developed a culture system to generate qHSC-like cells from human-induced pluripotent stem cells (hiPSCs) that can be converted into activated HSCs in culture. To monitor the activation process, a red fluorescent protein (RFP) gene was inserted in hiPSCs downstream of the activation marker gene actin alpha 2 smooth muscle (ACTA2). Using qHSC-like cells derived from RFP reporter iPSCs, we screened a repurposing chemical library and identified therapeutic candidates that prevent liver fibrosis. Hence, hiPSC-derived qHSC-like cells will be a useful tool to study the mechanism of HSC activation and to identify therapeutic agents.


Assuntos
Técnicas de Cultura de Células , Ciclo Celular , Descoberta de Drogas , Células Estreladas do Fígado/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Animais , Avaliação Pré-Clínica de Medicamentos , Perfilação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL
4.
APL Bioeng ; 5(2): 026104, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34027283

RESUMO

Interactions between the different liver cell types are critical to the maintenance or induction of their function in vitro. In this work, human-induced Pluripotent Stem Cells (hiPSCs)-derived Liver Sinusoidal Endothelial Cells (LSECs) and Hepatocytes-Like Cells (HLCs) were cultured and matured in a microfluidic environment. Both cell populations were differentiated in Petri dishes, detached, and inoculated in microfluidic biochips. In cocultures of both cell types, the tissue has exhibited a higher production of albumin (3.19 vs 5.31 µg/mL/106 cells in monocultures and cocultures) as well as a higher inducibility CYP450 over monocultures of HLCs. Tubular-like structures composed of LSECs and positive for the endothelial marker PECAM1, as well as a tissue more largely expressing Stabilin-2 were detected in cocultures only. In contrast, monocultures exhibited no network and less specific endothelial markers. The transcriptomic analysis did not reveal a marked difference between the profiles of both culture conditions. Nevertheless, the analysis allowed us to highlight different upstream regulators in cocultures (SP1, EBF1, and GATA3) and monocultures (PML, MECP2, and NRF1). In cocultures, the multi-omics dataset after 14 days of maturation in biochips has shown the activation of signaling related to hepatic maturation, angiogenesis, and tissue repair. In this condition, inflammatory signaling was also found to be reduced when compared to monocultures as illustrated by the activation of NFKB and by the detection of several cytokines involved in tissue injury in the latter. Finally, the extracted biological processes were discussed regarding the future development of a new generation of human in vitro hepatic models.

5.
NPJ Regen Med ; 6(1): 19, 2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33785758

RESUMO

Mesenchymal stromal cells (MSCs) are used for ameliorating liver fibrosis and aiding liver regeneration after cirrhosis; Here, we analyzed the therapeutic potential of small extracellular vesicles (sEVs) derived from interferon-γ (IFN-γ) pre-conditioned MSCs (γ-sEVs). γ-sEVs effectively induced anti-inflammatory macrophages with high motility and phagocytic abilities in vitro, while not preventing hepatic stellate cell (HSC; the major source of collagen fiber) activation in vitro. The proteome analysis of MSC-derived sEVs revealed anti-inflammatory macrophage inducible proteins (e.g., annexin-A1, lactotransferrin, and aminopeptidase N) upon IFN-γ stimulation. Furthermore, by enabling CX3CR1+ macrophage accumulation in the damaged area, γ-sEVs ameliorated inflammation and fibrosis in the cirrhosis mouse model more effectively than sEVs. Single cell RNA-Seq analysis revealed diverse effects, such as induction of anti-inflammatory macrophages and regulatory T cells, in the cirrhotic liver after γ-sEV administration. Overall, IFN-γ pre-conditioning altered sEVs resulted in efficient tissue repair indicating a new therapeutic strategy.

6.
Sci Rep ; 10(1): 14349, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32873852

RESUMO

Hepatocytes derived from human iPSCs are useful to study hepatitis B virus (HBV) infection, however infection efficiency is rather poor. In order to improve the efficiency of HBV infection to iPSC-derived hepatocytes, we set a co-culture of hepatocytes with liver non-parenchymal cells and found that liver sinusoidal endothelial cells (LSECs) enhanced HBV infection by secreting epidermal growth factor (EGF). While EGF receptor (EGFR) is known as a co-receptor for HBV, we found that EGF enhanced HBV infection at a low dose of EGF, whereas EGF at a high dose suppressed HBV infection. EGFR is internalized by clathrin-mediated endocytosis (CME) and clathrin-independent endocytosis (CIE) pathways depending on the dose of EGF. At a high dose of EGF, the endocytosed EGFR via CIE is degraded in the lysosome. This study is the first to provide evidence that HBV is endocytosed via CME and CIE pathways at a low and high dose of EGF, respectively. In conclusion, we developed an in vitro system of HBV infection using iPSC-derived liver cells, and show that EGF secreted from LSECs modulates HBV infection in a dose dependent manner.


Assuntos
Células Endoteliais/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Vírus da Hepatite B/metabolismo , Hepatite B/metabolismo , Fígado/citologia , Animais , Clatrina/metabolismo , Técnicas de Cocultura , Endocitose/efeitos dos fármacos , Endocitose/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Células Hep G2 , Hepatite B/virologia , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transfecção , Internalização do Vírus
7.
Mol Omics ; 16(2): 138-146, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31989141

RESUMO

Liver Sinusoidal Endothelial Cells (LSECs) are an important component of the liver as they compose the microvasculature which allows the supply of oxygen, blood, and nutrients. However, maintenance of these cells in vitro remains challenging as they tend to rapidly lose some of their characteristics such as fenestration or as their immortalized counterparts present poor characteristics. In this work, human induced pluripotent stem cells (hiPSCs) have been differentiated toward an LSEC phenotype. After differentiation, the RNA quantification allowed demonstration of high expression of specific vascular markers (CD31, CD144, and STAB2). Immunostaining performed on the cells was found to be positive for both Stabilin-1 and Stabilin-2. Whole transcriptome analysis performed with the nanoCAGE method further confirmed the overall vascular commitment of the cells. The gene expression profile revealed the upregulation of the APLN, LYVE1, VWF, ESAM and ANGPT2 genes while VEGFA appeared to be downregulated. Analysis of promoter motif activities highlighted several transcription factors (TFs) of interest in LSECs (IRF2, ERG, MEIS2, SPI1, IRF7, WRNIP1, HIC2, NFIX_NFIB, BATF, and PATZ1). Based on this investigation, we compiled the regulatory network involving the relevant TFs, their target genes as well as their related signaling pathways. The proposed hiPSC-derived LSEC model and its regulatory network were then confirmed by comparing the experimental data to primary human LSEC reference datasets. Thus, the presented model appears as a promising tool to generate more complex in vitro liver multi-cellular tissues.


Assuntos
Perfilação da Expressão Gênica/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/citologia , Diferenciação Celular , Células Cultivadas , Células Endoteliais/química , Células Endoteliais/citologia , Redes Reguladoras de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/química , Fígado/química , Nanotecnologia , Sequenciamento do Exoma
8.
Methods Mol Biol ; 1905: 131-142, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30536096

RESUMO

Human-induced pluripotent stem cells (iPSCs) could be a useful source for production of hepatocytes. Here, we develop protocols to generate iPSC-derived liver progenitor cells, liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs). We also establish long-term two-dimensional co-culture system to induce functional hepatocytes from iPSC-derived liver cells.


Assuntos
Técnicas de Cocultura/métodos , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Células Estreladas do Fígado/citologia , Humanos
9.
Stem Cell Reports ; 9(2): 490-498, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28757162

RESUMO

During liver development, hepatoblasts and liver non-parenchymal cells (NPCs) such as liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs) constitute the liver bud where they proliferate and differentiate. Accordingly, we reasoned that liver NPCs would support the maturation of hepatocytes derived from human induced pluripotent stem cells (hiPSCs), which usually exhibit limited functions. We found that the transforming growth factor ß and Rho signaling pathways, respectively, regulated the proliferation and maturation of LSEC and HSC progenitors isolated from mouse fetal livers. Based on these results, we have established culture systems to generate LSECs and HSCs from hiPSCs. These hiPSC-derived NPCs exhibited distinctive phenotypes and promoted self-renewal of hiPSC-derived liver progenitor cells (LPCs) over the long term in the two-dimensional culture system without exogenous cytokines and hepatic maturation of hiPSC-derived LPCs. Thus, a functional human liver model can be constructed in vitro from the LPCs, LSECs, and HSCs derived from hiPSCs.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/citologia , Molécula de Adesão de Leucócito Ativado/metabolismo , Animais , Biomarcadores , Diferenciação Celular/genética , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feto , Perfilação da Expressão Gênica , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Imunofenotipagem , Fígado/metabolismo , Regeneração Hepática , Camundongos , Fenótipo
10.
Stem Cell Reports ; 5(4): 508-15, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26365514

RESUMO

To develop a culture system for large-scale production of mature hepatocytes, liver progenitor cells (LPCs) with a high proliferation potential would be advantageous. We have found that carboxypeptidase M (CPM) is highly expressed in embryonic LPCs, hepatoblasts, while its expression is decreased along with hepatic maturation. Consistently, CPM expression was transiently induced during hepatic specification from human-induced pluripotent stem cells (hiPSCs). CPM(+) cells isolated from differentiated hiPSCs at the immature hepatocyte stage proliferated extensively in vitro and expressed a set of genes that were typical of hepatoblasts. Moreover, the CPM(+) cells exhibited a mature hepatocyte phenotype after induction of hepatic maturation and also underwent cholangiocytic differentiation in a three-dimensional culture system. These results indicated that hiPSC-derived CPM(+) cells share the characteristics of LPCs, with the potential to proliferate and differentiate bi-directionally. Thus, CPM is a useful marker for isolating hiPSC-derived LPCs, which allows development of a large-scale culture system for producing hepatocytes and cholangiocytes.


Assuntos
Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Metaloendopeptidases/análise , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Proteínas Ligadas por GPI/análise , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA