Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 19(1): 437, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930304

RESUMEN

BACKGROUND: Exosomes secreted from stem cells exerted salutary effects on the fibrotic liver. Herein, the roles of exosomes derived from human embryonic stem cell (hESC) in anti-fibrosis were extensively investigated. Compared with two-dimensional (2D) culture, the clinical and biological relevance of three-dimensional (3D) cell spheroids were greater because of their higher regeneration potential since they behave more like cells in vivo. In our study, exosomes derived from 3D human embryonic stem cells (hESC) spheroids and the monolayer (2D) hESCs were collected and compared the therapeutic potential for fibrotic liver in vitro and in vivo. RESULTS: In vitro, PKH26 labeled-hESC-Exosomes were shown to be internalized and integrated into TGFß-activated-LX2 cells, and reduced the expression of profibrogenic markers, thereby regulating cellular phenotypes. TPEF imaging indicated that PKH26-labeled-3D-hESC-Exsomes possessed an enhanced capacity to accumulate in the livers and exhibited more dramatic therapeutic potential in the injured livers of fibrosis mouse model. 3D-hESC-Exosomes decreased profibrogenic markers and liver injury markers, and improved the level of liver functioning proteins, eventually restoring liver function of fibrosis mice. miRNA array revealed a significant enrichment of miR-6766-3p in 3D-hESC-Exosomes, moreover, bioinformatics and dual luciferase reporter assay identified and confirmed the TGFßRII gene as the target of miR-6766-3p. Furthermore, the delivery of miR-6766-3p into activated-LX2 cells decreased cell proliferation, chemotaxis and profibrotic effects, and further investigation demonstrated that the expression of target gene TGFßRII and its downstream SMADs proteins, especially phosphorylated protein p-SMAD2/3 was also notably down-regulated by miR-6766-3p. These findings unveiled that miR-6766-3p in 3D-hESC-Exosomes inactivated SMADs signaling by inhibiting TGFßRII expression, consequently attenuating stellate cell activation and suppressing liver fibrosis. CONCLUSIONS: Our results showed that miR-6766-3p in the 3D-hESC-Exosomes inactivates smads signaling by restraining TGFßRII expression, attenuated LX2 cell activation and suppressed liver fibrosis, suggesting that 3D-hESC-Exosome enriched-miR-6766-3p is a novel anti-fibrotic therapeutics for treating chronic liver disease. These results also proposed a significant strategy that 3D-Exo could be used as natural nanoparticles to rescue liver injury via delivering antifibrotic miR-6766-3p.


Asunto(s)
Exosomas/metabolismo , Cirrosis Hepática/terapia , MicroARNs/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/metabolismo , Animales , Antagomirs/metabolismo , Técnicas de Cultivo Tridimensional de Células , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Exosomas/química , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología
2.
Liver Int ; 37(9): 1260-1271, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28135758

RESUMEN

The liver is a unique organ for homoeostasis with regenerative capacities. Hepatocytes possess a remarkable capacity to proliferate upon injury; however, in more severe scenarios liver regeneration is believed to arise from at least one, if not several facultative hepatic progenitor cell compartments. Newly identified pericentral stem/progenitor cells residing around the central vein is responsible for maintaining hepatocyte homoeostasis in the uninjured liver. In addition, hepatic progenitor cells have been reported to contribute to liver fibrosis and cancers. What drives liver homoeostasis, regeneration and diseases is determined by the physiological and pathological conditions, and especially the hepatic progenitor cell niches which influence the fate of hepatic progenitor cells. The hepatic progenitor cell niches are special microenvironments consisting of different cell types, releasing growth factors and cytokines and receiving signals, as well as the extracellular matrix (ECM) scaffold. The hepatic progenitor cell niches maintain and regulate stem cells to ensure organ homoeostasis and regeneration. In recent studies, more evidence has been shown that hepatic cells such as hepatocytes, cholangiocytes or myofibroblasts can be induced to be oval cell-like state through transitions under some circumstance, those transitional cell types as potential liver-resident progenitor cells play important roles in liver pathophysiology. In this review, we describe and update recent advances in the diversity and plasticity of hepatic progenitor cell and their niches and discuss evidence supporting their roles in liver homoeostasis, regeneration, fibrosis and cancers.


Asunto(s)
Regeneración Hepática , Hígado/citología , Células Madre/citología , Animales , Comunicación Celular , Proliferación Celular , Hematopoyesis , Hepatocitos/citología , Homeostasis , Humanos , Hígado/patología , Modelos Biológicos , Transducción de Señal
3.
Lab Invest ; 96(3): 350-60, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26692291

RESUMEN

Although hepatic progenitor cells (HPCs) are known to contribute to cholestatic liver fibrosis (CLF), how Notch signaling modulates the differentiation of HPCs to cholangiocytes in CLF is unknown. Thus, using a rat model of CLF that is induced by bile duct ligation, we inhibited Notch signaling with DAPT. In vivo, CK19, OV6, Sox9, and EpCAM expression was increased significantly. Notch signaling increased after bile duct ligation, and DAPT treatment reduced the expression of CK19, OV6, Sox9, and EpCAM and blocked cholangiocyte proliferation and CLF. In vitro, treatment of a WB-F344 cell line with sodium butyrate resulted in increased mRNA and protein expression of CK19, Sox9, and EpCAM, but Notch signaling was activated. Both of these processes were inhibited by DAPT. This study reveals that Notch signaling activation is required for HPC differentiation into cholangiocytes in CLF, and inhibition of the Notch signaling pathway may offer a therapeutic approach for treating CLF.


Asunto(s)
Colestasis/complicaciones , Cirrosis Hepática/etiología , Hígado/citología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Células Madre/citología , Animales , Diferenciación Celular , Cirrosis Hepática/prevención & control , Ratas , Ratas Sprague-Dawley
4.
Stem Cells ; 31(10): 2095-103, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836547

RESUMEN

Human embryonic stem cells (hESCs) can be progressively differentiated into definitive endoderm (DE), hepatic progenitors, and hepatocytes, and thus provide an excellent model system for the mechanistic study of hepatocyte differentiation, which is currently poorly understood. Here, we found that insulin enhanced hepatocyte differentiation from hESC-derived DE. Insulin activated the PI3K/AKT pathway, but not the mitogen-activated protein kinase pathway in the DE cells, and inhibition of the PI3K/AKT pathways by inhibitors markedly inhibited hepatocyte differentiation. In addition, insulin-like growth factor 1 (IGF1) and IGF2 also activated the PI3K/AKT pathway in DE cells and their expression was robustly upregulated during hepatocyte differentiation from DE. Furthermore, inhibition of IGF receptor 1 (IGF1R) by a small molecule inhibitor PPP or knockdown of the IGF1R by shRNA attenuated hepatocyte differentiation. Moreover, simultaneous knockdown of the IGF1R and the insulin receptor with shRNAs markedly reduced the activation of AKT and substantially impaired hepatocyte differentiation. The PI3K pathway specifically enhanced the expression of HNF1 and HNF4 to regulate hepatocyte differentiation from DE. Although inhibition of the PI3K pathway was previously shown to be required for the induction of DE from hESCs, our study revealed a positive role of the PI3K pathway in hepatocyte differentiation after the DE stage, and has advanced our understanding of hepatocyte cell fate determination.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Hepatocitos/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Insulina/fisiología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Línea Celular , Supervivencia Celular , Humanos , Factor II del Crecimiento Similar a la Insulina/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/fisiología , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
5.
Stem Cell Res Ther ; 13(1): 218, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35619172

RESUMEN

BACKGROUND: Human pluripotent stem cells (hPSCs) have great potential in applications for regenerative medicine and drug development. However, 3D suspension culture systems for clinical-grade hPSC large-scale production have been a major challenge. Accumulating evidence has demonstrated that the addition of dextran sulfate (DS) could prevent excessive adhesion of hPSCs from forming larger aggregates in 3D suspension culture. However, the signaling and molecular mechanisms underlying this phenomenon remain elusive. METHODS: By using a cell aggregate culture assay and separating big and small aggregates in suspension culture systems, the potential mechanism and downstream target genes of DS were investigated by mRNA sequence analysis, qRT-PCR validation, colony formation assay, and interference assay. RESULTS: Since cellular adhesion molecules (CAMs) play important roles in hPSC adhesion and aggregation, we assumed that DS might prevent excess adhesion through affecting the expression of CAMs in hPSCs. As expected, after DS treatment, we found that the expression of CAMs was significantly down-regulated, especially E-cadherin (E-cad) and intercellular adhesion molecule 1 (ICAM1), two highly expressed CAMs in hPSCs. The role of E-cad in the adhesion of hPSCs has been widely investigated, but the function of ICAM1 in hPSCs is hardly understood. In the present study, we demonstrated that ICAM1 exhibited the capacity to promote the adhesion in hPSCs, and this adhesion was suppressed by the treatment with DS. Furthermore, transcriptomic analysis of RNA-seq revealed that DS treatment up-regulated genes related to Wnt signaling resulting in the activation of Wnt signaling in which SLUG, TWIST, and MMP3/7 were highly expressed, and further inhibited the expression of E-cad. CONCLUSION: Our results demonstrated that DS played an important role in controlling the size of hPSC aggregates in 3D suspension culture by inhibiting the expression of ICAM1 coupled with the down-regulation of E-cad through the activation of the Wnt signaling pathway. These results represent a significant step toward developing the expansion of hPSCs under 3D suspension condition in large-scale cultures.


Asunto(s)
Células Madre Pluripotentes , Vía de Señalización Wnt , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular , Sulfato de Dextran , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Células Madre Pluripotentes/metabolismo
6.
J Hepatol ; 55(4): 838-45, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21334406

RESUMEN

BACKGROUND & AIMS: Our previous studies showed that CD133, EpCAM, and aldehyde dehydrogenase (ALDH) are useful markers to identify cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) tissues. The present study aims to evaluate chemosensitivity and invasion capability of HCC based on CSC marker profiles, and to explore the underlying molecular mechanisms. METHODS: Hepatoma cell lines were separated into subpopulations according to CD133, EpCAM, and ALDH expression profiles. Epithelial mesenchymal transition (EMT) and hedgehog (Hh) signaling were examined to identify their links with chemoresistance and aggressive invasion. RESULTS: Well-differentiated cell lines were positive for CD133(+)/ALDH(high) and CD133(+)/EpCAM(+) at 1.5-15% and 2.3-8.3%; whereas, poorly-differentiated cells were almost all negative for these markers. FACS-enriched CD133(+)/ALDH(high) and CD133(+)/EpCAM(+) Hep3B and Huh-7 cells formed more spheroids in vitro. CD133(-)/ALDH(low) HLE cells were more resistant to cisplatin, doxorubicin or sorafenib than their positive counterparts. CD133(-)/EpCAM(-) Huh-7 cells or CD133(-)/ALDH(-) HLE cells exhibited a higher invasion rate than their positive counterparts. HLE and HLF cells acquired EMT in double negative subpopulations. Hh activity in Huh-7 CD133(-)/EpCAM(-) cells was higher than in their positive counterparts, and the inhibition of Hh activity by cyclopamine resulted in reduced cell proliferation. CONCLUSIONS: Well-differentiated CD133(+)/ALDH(high) or CD133(+)/EpCAM(+) cells appear to be a CSC/initiating subpopulation; whereas, in poorly-differentiated hepatoma cells, EMT and enhanced hedgehog signaling activity may be responsible for their chemoresistance and invasion. These findings underscore the significance of EMT and enhanced Hh signaling in liver cancer stem or initiating cells.


Asunto(s)
Carcinoma Hepatocelular , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/fisiología , Proteínas Hedgehog/metabolismo , Neoplasias Hepáticas , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/fisiopatología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Citometría de Flujo , Glicoproteínas/metabolismo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/fisiopatología , Ratones , Invasividad Neoplásica , Péptidos/metabolismo , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochem Biophys Res Commun ; 407(2): 295-300, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21382341

RESUMEN

The capacity of stem cells to differentiate into specific cell types makes them very promising in tissue regeneration and repair. However, realizing this promise requires novel methods for guiding lineage-specific differentiation of stem cells. In this study, hepatocyte growth factor (HGF), an important morphogen in liver development, was co-printed with collagen I (Col) to create arrays of protein spots on glass. Human adipose stem cells (ASCs) were cultured on top of the HGF/Col spots for 2weeks. The effects of surface-immobilized HGF on hepatic differentiation of ASCs were analyzed using RT-PCR, ELISA and immunocytochemistry. Stimulation of stem cells with HGF from the bottom-up caused an upregulation in synthesis of α-fetoprotein and albumin, as determined by immunocytochemistry and ELISA. RT-PCR results showed that the mRNA levels for albumin, α-fetoprotein and α1-antitrypsin were 10- to 20-fold higher in stem cells cultured on the HGF/Col arrays compared to stem cells on Col only spots. Our results show that surfaces containing HGF co-printed with ECM proteins may be used to differentiate mesenchymal stem cells such as ASCs into hepatocyte-like cells. These results underscore the utility of growth factor-containing culture surfaces for stem cell differentiation.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/citología , Proteínas Inmovilizadas/farmacología , Regeneración Hepática , Células Madre Mesenquimatosas/efectos de los fármacos , Tejido Adiposo/citología , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Colágeno Tipo I/metabolismo , Expresión Génica , Humanos , Hígado/citología , Hígado/metabolismo
8.
Liver Transpl ; 17(4): 418-27, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21445925

RESUMEN

The transplantation of primary hepatocytes has been shown to augment the function of damaged livers and to bridge patients to liver transplantation. However, primary hepatocytes often have low levels of engraftment and survive for only a short time after transplantation. To explore the potential benefits of using decellularized liver matrix (DLM) as a carrier for hepatocyte transplantation, DLM from whole mouse livers was generated. Human fetal hepatocytes immortalized by telomerase reconstitution (FH-hTERTs) or primary human hepatocytes were infused into the DLM, which was then implanted into the omenta of immunodeficient nonobese diabetic/severe combined immunodeficient/interleukin-2 receptor γ-deficient mice or nonobese diabetic/severe combined immunodeficient/mucopolysaccharidosis type VII mice. The removal of endogenous cellular components and the preservation of the extracellular matrix proteins and vasculature were demonstrated in the resulting DLM. Bioluminescent imaging revealed that FH-hTERTs transduced with a lentiviral vector expressing firefly luciferase survived in the DLM for 8 weeks after peritoneal implantation, whereas the luciferase signal from FH-hTERTs rapidly declined in control mice 3 to 4 weeks after transplantation via splenic injection or omental implantation after Matrigel encapsulation. Furthermore, primary human hepatocytes that were reconstituted in the DLM not only survived 6 weeks after transplantation but also maintained their function, as demonstrated by messenger RNA levels of albumin and cytochrome P450 (CYP) subtypes (CYP3A4, CYP2C9, and CYP1A1) similar to the levels in freshly isolated human primary hepatocytes (hPHs). In contrast, when hPHs were transplanted into mice via splenic injection, they failed to express CYP3A4, although they expressed albumin. In conclusion, DLM provides an excellent environment for long-term survival and maintenance of the hepatocyte phenotype after transplantation.


Asunto(s)
Trasplante de Tejido Fetal/métodos , Hepatocitos/trasplante , Hígado/citología , Animales , Supervivencia Celular , Células Cultivadas , Hepatocitos/fisiología , Humanos , Ratones
9.
Stem Cells ; 28(4): 674-86, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20135682

RESUMEN

Human embryonic stem cells (hESCs) may provide a cell source for functional hepatocytes for clinical applications and drug development. Initially, the hESC population was enriched to be more than 85% definitive endoderm (DE) as assessed by the expression of CXCR4, SOX17, and FOXA2. We then successfully converted DE into hepatic progenitors with 93% of the cells being positive for alpha-feto protein within 9 days. The percentage of albumin positive cells gradually increased to 90% at days 20-22 after differentiation. Moreover, our hESC-derived hepatocytes (hEH) developed a complete biotransformation system including phase I and II metabolizing enyzmes and phase III transporters. Nuclear receptors, which are critical in regulating the expression of metabolizing enzymes, were also expressed by our hEH. Using ultraperformance liquid chromatography-tandem mass spectrometry technology, we identified seven metabolic pathways of the drug bufuralol including four newly-reported ones in our hEH, which are the same as those in freshly isolated human primary hepatocytes (hPH). In addition, the results of the metabolism of four drugs indicate that our hEH have the capacity to metabolize these drugs at levels that are comparable to hPH. In conclusion, we have generated a relatively homogenous population of hepatocytes from hESCs, which appear to have complete metabolic function that is comparable to primary liver cells. These results represent a significant step towards the efficient differentiation of mature hepatocytes for cell-based therapeutics as well as for pharmacology and toxicology studies.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Humanos
10.
Cell Prolif ; 54(9): e13112, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34390064

RESUMEN

OBJECTIVES: For clinical applications of cell-based therapies, a large quantity of human pluripotent stem cells (hPSCs) produced in standardized and scalable culture processes is required. Currently, microcarrier-free suspension culture shows potential for large-scale expansion of hPSCs; however, hPSCs tend to aggregate during culturing leading to a negative effect on cell yield. To overcome this problem, we developed a novel protocol to effectively control the sizes of cell aggregates and enhance the cell proliferation during the expansion of hPSCs in suspension. MATERIALS AND METHODS: hPSCs were expanded in suspension culture supplemented with polyvinyl alcohol (PVA) and dextran sulphate (DS), and 3D suspension culture of hPSCs formed cell aggregates under static or dynamic conditions. The sizes of cell aggregates and the cell proliferation as well as the pluripotency of hPSCs after expansion were assessed using cell counting, size analysis, real-time quantitative polymerase chain reaction, flow cytometry analysis, immunofluorescence staining, embryoid body formation, teratoma formation and transcriptome sequencing. RESULTS: Our results demonstrated that the addition of DS alone effectively prevented hPSC aggregation, while the addition of PVA significantly enhanced hPSC proliferation. The combination of PVA and DS not only promoted cell proliferation of hPSCs but also produced uniform and size-controlled cell aggregates. Moreover, hPSCs treated with PVA, or DS or a combination, maintained the pluripotency and were capable of differentiating into all three germ layers. mRNA-seq analysis demonstrated that the combination of PVA and DS significantly promoted hPSC proliferation and prevented cell aggregation through improving energy metabolism-related processes, regulating cell growth, cell proliferation and cell division, as well as reducing the adhesion among hPSC aggregates by affecting expression of genes related to cell adhesion. CONCLUSIONS: Our results represent a significant step towards developing a simple and robust approach for the expansion of hPSCs in large scale.


Asunto(s)
Agregación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sulfato de Dextran/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Alcohol Polivinílico/farmacología , Animales , Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ratones
11.
Am J Physiol Gastrointest Liver Physiol ; 298(1): G101-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19892940

RESUMEN

Activation of hepatic stellate cells (HSC) results in their proliferation and in the secretion of extracellular matrix (ECM) proteins, which leads to hepatic fibrosis. microRNAs (miRNAs) have been shown to regulate various cell functions, such as proliferation, differentiation, and apoptosis. Hence, we have analyzed the miRNAs that were differentially expressed in HSC isolated from sham-operated and bile duct-ligated rats. Expression of two miRNAs, miRNA-150 and miRNA-194, was reduced in HSC isolated from fibrotic rats compared with sham-operated animals. These two miRNAs were overexpressed in LX-2 cells, and their ability to inhibit cell proliferation, the expression of smooth muscle alpha-actin (SMA), a marker for activation, and collagen type I, a marker for ECM secretion, was determined. Overexpression of these two miRNAs resulted in a significant inhibition of proliferation (P < 0.05) and reduced SMA and collagen I levels compared with either untreated cells or nonspecific miRNA-expressing cells. Next, the protein targets of these two miRNAs were found using bioinformatics approaches. C-myb was found to be a target for miRNA-150, and rac 1 was found to be one of the targets for miRNA-194. Therefore, we studied the expression of these two proteins by overexpressing these two miRNAs in LX-2 cells and found that overexpression of miRNA-150 and miRNA-194 resulted in a significant inhibition of c-myb and rac 1 expression, respectively. We conclude that both miRNA-150 and miRNA-194 inhibit HSC activation and ECM production, at least in part, via inhibition of c-myb and rac 1 expression.


Asunto(s)
Matriz Extracelular/fisiología , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática , MicroARNs/genética , MicroARNs/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Secuencia de Bases , Línea Celular Transformada , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Bases de Datos Genéticas , Regulación hacia Abajo/genética , Expresión Génica/fisiología , Genes myb/genética , Células Estrelladas Hepáticas/citología , Humanos , Etiquetado Corte-Fin in Situ , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Masculino , MicroARNs/aislamiento & purificación , Neuropéptidos/genética , Neuropéptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
12.
Gastroenterology ; 136(5): 1783-95.e10, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19208340

RESUMEN

BACKGROUND & AIMS: Despite high morbidity and mortality of alcoholic liver disease worldwide, the molecular mechanisms underlying alcohol-induced liver cell death are not fully understood. Transglutaminase 2 (TG2) is a cross-linking enzyme implicated in apoptosis. TG2 levels and activity are increased in association with various types of liver injury. However, how TG2 induces hepatic apoptosis is not known. METHODS: Human hepatic cells or primary hepatocytes from rats or TG2+/+ and TG2-/- mice were treated with ethanol. Mice were administered anti-Fas antibody or alcohol. Liver sections were prepared from patients with alcoholic steatohepatitis. Changes in TG2 levels, Sp1 cross-linking and its activities, expression of hepatocyte growth factor receptor, c-Met, and hepatic apoptosis were measured. RESULTS: Ethanol induced apoptosis in hepatic cells, enhanced activity and nuclear accumulation of TG2 as well as accumulation of cross-linked and inactivated Sp1, and reduced expression of the Sp1-responsive gene, c-Met. These effects were rescued by TG2 knockdown, restoration of functional Sp1, or addition of hepatocyte growth factor, whereas apoptosis was reproduced by Sp1 knockdown or TG2 overexpression. Compared with TG2+/+ mice, TG2-/- mice showed markedly reduced hepatocyte apoptosis and Sp1 cross-linking following ethanol or anti-Fas treatment. Treatment of TG2+/+ mice with the TG2 inhibitors putrescine or cystamine blocked anti-Fas-induced hepatic apoptosis and Sp1 silencing. Moreover, enhanced expression of cross-linked Sp1 and TG2 was evident in hepatocyte nuclei of patients with alcoholic steatohepatitis. CONCLUSIONS: TG2 induces hepatocyte apoptosis via Sp1 cross-linking and inactivation, with resultant inhibition of the expression of c-Met required for hepatic cell viability.


Asunto(s)
Etanol/farmacología , Hígado Graso Alcohólico/metabolismo , Proteínas de Unión al GTP/metabolismo , Silenciador del Gen , Factor de Transcripción Sp1/metabolismo , Transglutaminasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Hígado Graso Alcohólico/patología , Cobayas , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción Sp1/genética , Activación Transcripcional , Transfección
13.
Liver Int ; 30(3): 376-87, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20015148

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) have emerged as novel genetic regulators of cell functions such as proliferation, apoptosis and cancer. AIMS: The aim of this study was to evaluate the role of a specific miRNA in modulating hepatic cell functions. METHODS: C57Bl/6 mice were administered anti-fas receptor antibodies to induce liver cell apoptosis. miRNAs were purified from the liver tissue and evaluated using an miRNA microarray. The role of miRNA-491_5p, which was overexpressed in the model, in modulating hepatic cell functions was evaluated. miRNA-491_5p was overexpressed in Hep G2 cells, followed by the addition of tumour necrosis factor (TNF)-alpha, and induction of apoptosis as well as genes involved in apoptosis pathways were evaluated. The effect of miRNA-491_5p target genes on apoptosis was also analysed by inhibiting their expression by siRNA-induced gene silencing. RESULTS: Upregulation of miRNA-491_5p was found in a high-dose anti-fas receptor antibody group. Overexpression of microRNA-491_5p sensitized Hep G2 cells for TNF-alpha-induced apoptosis, and also caused an inhibition of alpha-fetoprotein, (AFP), heat shock protein-90 (hsp-90) and nuclear factor-kappaB (NF-kappaB). Overexpression of miRNA-491_5p or inhibition of AFP and hsp-90 resulted in an increased apoptosis in TNF-alpha-treated Hep G2 cells. CONCLUSIONS: One of the miRNAs that is associated with the acute liver injury mouse model, miRNA-491_5p, sensitizes Hep G2 cells for TNF-alpha-induced apoptosis, at least in part, by inhibiting AFP, hsp-90 and NF-kappaB.


Asunto(s)
Apoptosis/efectos de los fármacos , Hepatopatías/metabolismo , Hígado/metabolismo , MicroARNs/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Enfermedad Aguda , Animales , Anticuerpos/farmacología , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Células Hep G2 , Humanos , Hígado/patología , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba , Receptor fas/inmunología
14.
Exp Mol Pathol ; 89(1): 27-35, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20511115

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) are thought to be a critical subpopulation in tumor development, progression, metastasis and recurrence, and the identification of these cells is an initial step in understanding their role in oncogenesis and in seeking valuable markers for diagnosis or development of targeting therapeutics. AIMS: To identify CSCs in hepatocellular carcinoma (HCC) specimens and define their tissue specificity. METHODS: Immunohistochemical staining of CSC markers: CD44, CD90, CD133 and aldehyde dehydrogenase (ALDH) was performed in 25 HCC specimens, 4 hepatoblastomas, 8 peri-malignant tissues, and 19 cases of viral hepatitis. RESULTS: The positivity of CD44 staining in HCC specimens was significantly lower than in viral hepatitis specimens. The positive rate of CD133 in HCC was similar to viral hepatitis specimens. CD133(+) cells were largely localized to ALDH-positive cells in HCC as revealed by confocal microscopy. In contrast, the co-expression of both markers was visualized within vessels or in the portal areas in viral hepatitis. Moreover, among 7 liver specimens adjacent to HCC tissue, 3-6 samples were positive for CD44, CD90, CD133 and ALDH, especially in dysplastic cells. One of 4 hepatoblastoma cases was positive for all these markers; whereas, the other three specimens were negative for all these CSC markers. CONCLUSIONS: In HCC and dysplastic tissues, clusters of CD133(+)/ALDH(high) cells were identified. The use of cancer stem cell markers to screen tissues with chronic liver diseases provides limited guidance in the identification of malignant cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Hepatitis Viral Humana/metabolismo , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/metabolismo , Antígeno AC133 , Adulto , Anciano , Aldehído Deshidrogenasa/metabolismo , Antígenos CD/metabolismo , Carcinoma Hepatocelular/patología , Femenino , Glicoproteínas/metabolismo , Hepatitis Viral Humana/patología , Humanos , Receptores de Hialuranos/metabolismo , Técnicas para Inmunoenzimas , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Péptidos/metabolismo , Antígenos Thy-1/metabolismo , Adulto Joven
15.
Biochem Biophys Res Commun ; 379(4): 876-81, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19133235

RESUMEN

One of the pathways by which alcohol induces hepatocyte apoptosis is via oxidative stress. We screened several chemically-synthesized small molecules and found LAS-0811, which inhibits oxidative stress. In this study, we elucidated its role in inhibiting alcohol-induced apoptosis in hepatocyte-like VL-17A cells. VL-17A cells were pre-incubated with LAS-0811, followed by ethanol incubation. Ethanol-induced reactive oxygen species and apoptosis were significantly inhibited in LAS-0811 pre-treated cells. VL-17A cells were transfected with a reporter (ARE/TK-GFP) plasmid containing green fluorescent protein (GFP) as a reporter gene and the anti-oxidant response element as the promoter. LAS-0811 pre-treatment significantly induced the GFP expression compared to the cells treated with ethanol alone. LAS-0811 induced the activation of nrf2 and enhanced the expression and activity of glutathione peroxidase, one of the downstream targets of nrf2. The results indicate that LAS-0811 protects VL-17A cells against ethanol-induced oxidative stress and apoptosis at least in part via nrf2 activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Citoprotección , Etanol/antagonistas & inhibidores , Hepatocitos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Línea Celular , Etanol/toxicidad , Genes Reporteros/efectos de los fármacos , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Elementos de Respuesta/efectos de los fármacos
16.
Anal Chem ; 80(16): 6351-7, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18642875

RESUMEN

The microarray format is being used extensively for combinatorial screening of cellular interactions with proteins, small molecules, or biomaterials. The utility of microarray-based cell cultivation approaches may be enhanced further by incorporating biosensing elements alongside the cell-adhesive ligands to enable local detection of secreted cellular products. The concept of combining cells and sensing elements in the same microarray is demonstrated in the present paper with hepatocytes serving as a model cellular system. Robotic microarraying was employed to print arrays of 300-mum-diameter collagen (I) spots alongside the antibody (Ab) spots specific to liver proteins: albumin and alpha1-antitrypsin (alpha1-AT). Protein microarrays were printed onto poly(ethylene glycol) hydrogel-coated glass slides, thus eliminating nonspecific adsorption of cells or proteins. When incubated with printed microarrays, hepatocytes became localized on collagen (I) domains but did not attach on Ab spots or elsewhere on hydrogel-coated glass substrates. Liver-specific proteins secreted by hepatocytes were captured on Ab domains in the immediate vicinity of the cells, detected with a sandwich immunofluorescent assay and quantified using a microarray scanner. Importantly, hepatic albumin and alpha1-AT production detected in the microarray was comparable to enzyme-linked immunosorbent assay measurements of these proteins. In the future, the juxtaposition of sensing Ab regions with cell arrays will be particularly useful for the detection of local appearance or loss of phenotype of cells interacting with the printed components of the cellular microenvironment.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Hepatocitos/metabolismo , Polietilenglicoles/química , Análisis por Matrices de Proteínas/métodos , Albúminas/inmunología , Albúminas/metabolismo , Animales , Carcinoma Hepatocelular/inmunología , Células Cultivadas , Colágeno Tipo I/inmunología , Colágeno Tipo I/metabolismo , Ensayo de Inmunoadsorción Enzimática , Vidrio/química , Hepatocitos/citología , Humanos , Hidrogeles/química , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Ratas , alfa 1-Antitripsina/inmunología , alfa 1-Antitripsina/metabolismo
17.
Stem Cells ; 25(12): 3058-68, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17885076

RESUMEN

Human embryonic stem cells (hESC) may provide a cell source for functional hepatocytes. The aim of this study is to establish a viable human hepatocyte-like cell line from hESC that can be used for cell-based therapies. The differentiated hESC were enriched by transducing with a lentivirus vector containing the green fluorescent protein (GFP) gene driven by the alpha1-antitrypsin promoter; the GFP gene is expressed in committed hepatocyte progenitors and hepatocytes. GFP+ hESC were purified by laser microdissection and pressure catapulting. In addition, differentiated hESC that were transduced with a lentivirus triple-fusion vector were transplanted into NOD-SCID mice, and the luciferase-induced bioluminescence in the livers was evaluated by a charge-coupled device camera. GFP+ hESC expressed a large series of liver-specific genes, and expression levels of these genes were significantly improved by purifying GFP+ hESC; our results demonstrated that purified differentiated hESC express nearly physiological levels of liver-specific genes and have liver-specific functions that are comparable to those of primary human hepatocytes. The differentiated hESC survived and engrafted in mouse livers, and human liver-specific mRNA and protein species were detected in the transplanted mouse liver and serum at 3 weeks after transplantation. This is the first time that human albumin generated by hESC-derived hepatocytes was detected in the serum of an animal model. This also represents the first successful transplantation of differentiated hESC in an animal liver and the first bioluminescence imaging of hESC in the liver. This study is an initial step in establishing a viable hepatocyte-like cell line from hESC. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Hepatocitos/fisiología , Hepatocitos/trasplante , Animales , Carcinoma Hepatocelular/patología , Diferenciación Celular/genética , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Supervivencia de Injerto , Hepatocitos/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID
18.
Cell Transplant ; 17(8): 899-909, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19069633

RESUMEN

Noninvasive serial monitoring of the fate of transplanted cells would be invaluable to evaluate the potential therapeutic use of human hepatocyte transplantation. Therefore, we assessed the feasibility of bioluminescent imaging using double or triple fusion lentiviral vectors in a NOD-SCID mouse model transplanted with immortalized human fetal hepatocytes. Lentiviral vectors driven by the CMV promoter were constructed carrying reporter genes: firefly luciferase and green fluorescence protein with or without herpes simplex virus type 1 thymidine kinase. Human fetal hepatocytes immortalized by telomerase reconstitution (FH-hTERT) were successfully transduced with either of these fusion vectors. Two million stably transduced cells selected by fluorescence-activated cell sorting were injected into the spleens of NOD-SCID mice pretreated with methylcholanthrene and monocrotaline. The transplanted mice were serially imaged with a bioluminescence charged-coupled device camera after D-luciferin injection. Bioluminescence signal intensity was highest on day 3 (6.10 +/- 2.02 x 10(5) p/s/cm2/sr, mean +/- SEM), but decreased to 2.26 +/- 1.54 x 10(5) and 7.47 +/- 3.09 x 10(4) p/s/cm2/sr on day 7 and 10, respectively (p = 0.001). ELISA for human albumin in mice sera showed that levels were similar to those of control mice on day 2 (3.25 +/- 0.92 vs. 2.84 +/- 0.59 ng/ml, mean +/- SEM), peaked at 18.04 +/- 3.11 ng/ml on day 7, and decreased to 8.93 +/- 1.40 and 3.54 +/- 0.87 ng/ml on day 14 and 21, respectively (p = 0.02). Real-time quantitative RT-PCR showed gene expression levels of human albumin, alpha1-antitrypsin, and transferrin in mouse liver were 60.7 +/- 6.5%, 26.0 +/- 1.4%, and 156.8 +/- 62.4% of those of primary human adult hepatocytes, respectively, and immunohistochemistry revealed cells with human albumin and alpha1-antitrypsin expression in the mouse liver. In conclusion, our study demonstrated that bioluminescent imaging appears to be a sensitive, noninvasive modality for serial monitoring of transplanted hepatic stem cells.


Asunto(s)
Células Madre Fetales/trasplante , Hepatocitos/trasplante , Mediciones Luminiscentes/métodos , Trasplante de Células Madre/métodos , Albúminas/genética , Animales , Carcinógenos/farmacología , Línea Celular Transformada , Modelos Animales de Enfermedad , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Genes Reporteros/genética , Vectores Genéticos/genética , Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes/genética , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Lentivirus/genética , Luciferasas de Luciérnaga/genética , Ratones , Ratones SCID , Bazo/citología , Bazo/cirugía , Transfección/métodos , Transferrina/genética , alfa 1-Antitripsina/genética
19.
Cell Signal ; 19(11): 2339-50, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17728104

RESUMEN

UNLABELLED: Ethanol abuse is one of the major etiologies of cirrhosis. Ethanol has been shown to induce apoptosis via activation of oxidative stress, mitogen-activated protein kinases (MAPK), and tyrosine kinases. However, there is a paucity of data that examine the interplay among these molecules. In the present study we have systematically elucidated the role of novel protein kinase C isoforms (nPKC; PKCdelta and PKCepsilon) in ethanol-induced apoptosis in hepatocytes. Ethanol enhanced membrane translocation of PKCdelta and PKCepsilon, which was associated with the phosphorylation of p38MAPK, p42/44MAPK and JNK1/2, and the nuclear translocation of NF-kappaB and AP-1. This resulted in increased apoptosis in primary rat hepatocytes. Inhibition of both PKCdelta and PKCepsilon resulted in a decreased MAPK activation, decreased nuclear translocation of NF-kappaB and AP-1, and inhibition of apoptosis. In addition, ethanol activated the tyrosine phosphorylation of PKCdelta via tyrosine kinase in hepatocytes. The tyrosine phosphorylated PKCdelta was cleaved by caspase-3 and these fragments were translocated to the nucleus. Inhibition of ethanol-induced oxidative stress blocked the membrane translocation of PKCdelta and PKCepsilon, and the tyrosine phosphorylation of PKCdelta in hepatocytes. Inhibition of oxidative stress, tyrosine kinase or caspase-3 activity caused a decreased nuclear translocation of PKCdelta in response to ethanol, and was associated with less apoptosis. CONCLUSION: These results provide a newly-described mechanism by which ethanol induces apoptosis via activation of nPKC isoforms in hepatocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/farmacología , Hepatocitos/citología , Hepatocitos/enzimología , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Caspasa 3/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Fluoresceínas/farmacología , Hepatocitos/efectos de los fármacos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Masculino , Metaloporfirinas/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Polietilenglicoles/farmacología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/farmacología
20.
Stem Cells Dev ; 27(4): 252-261, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29256825

RESUMEN

Hepatocytes differentiated from human embryonic stem cells (ESCs) could provide a powerful tool for enabling cell-based therapies, studying the mechanisms underlying human liver development and disease, and testing the efficacy and safety of pharmaceuticals. However, currently most in vitro protocols yield hepatocytes with low levels of liver function. In this study, we investigated the potential of Salvianolic acid B (Sal B), an active pharmaceutical compound present in Salvia miltiorrhiza, which has been shown to have an antifibrotic effect in previous studies, to enhance hepatocyte differentiation from human ESCs. After treatment with Sal B, albumin expression and secretion were consistently increased, indicating that Sal B could promote hepatocyte differentiation process. Expression of a large number of important phase 1 and 2 metabolizing enzymes and phase 3 transporters was also increased in treated cells, indicating an enhanced biotransformation function. Our investigations further revealed the activation of Wnt pathway in treated cells, as determined by upregulation of Wnts, which increased amounts of nuclear ß-catenin. This increased nuclear ß-catenin led in turn to the enhanced expression of T cell factor (TCF) 3 and lymphoid enhancer-binding factor (LEF) 1 which upregulated their downstream targets, cyclin D1 and c-Myc. Notch receptors (Notch1, Notch3), Notch ligand (Jagged2), and Notch receptor targets [hairy and enhancer of split (Hes) 1, 5] were downregulated in treated cells, suggesting that Notch pathway was inhibited. Consistent with the inhibition of Notch pathway, expression of cholangiocyte marker, CK7, was significantly reduced by treatment with Sal B. Numb, a direct transcriptional target of Wnt pathway and a negative regulator of Notch pathway, was upregulated, consistent with activation of Wnt signaling and suppression of Notch signaling. In conclusion, our study demonstrated that Sal B enhanced hepatocyte differentiation from human ESCs through activation of Wnt pathway and inhibition of Notch pathway. Therefore, this study suggests that Sal B can be used as a potential agent to generate more mature hepatocytes for cell-based therapeutics and pharmaceutical studies.


Asunto(s)
Benzofuranos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Receptores Notch/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Células Cultivadas , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Hígado/citología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA