Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Biol Med (Maywood) ; 244(8): 702-708, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30803263
2.
Stem Cells ; 37(3): 306-317, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30471152

RESUMEN

Directed differentiation of human induced pluripotent stem cells (iPSCs) toward hepatobiliary lineages has been increasingly used as models of human liver development/diseases. As protein kinases are important components of signaling pathways regulating cell fate changes, we sought to define the key molecular mediators regulating human liver development using inhibitors targeting tyrosine kinases during hepatic differentiation of human iPSCs. A library of tyrosine kinase inhibitors was used for initial screening during the multistage differentiation of human iPSCs to hepatic lineage. Among the 80 kinase inhibitors tested, only Src inhibitors suppressed endoderm formation while none had significant effect on later stages of hepatic differentiation. Transient inhibition of c-Src during endodermal induction of human iPSCs reduced endodermal commitment and expression of endodermal markers, including SOX17 and FOXA2, in a dose-dependent manner. Interestingly, the transiently treated cells later developed into profibrogenic cholangiocyte-like cells expressing both cholangiocyte markers, such as CK7 and CK19, and fibrosis markers, including Collagen1 and smooth muscle actin. Further analysis of these cells revealed colocalized expression of collagen and yes-associated protein (YAP; a marker associated with bile duct proliferation/fibrosis) and an increased production of interleukin-6 and tumor necrosis factor-α. Moreover, treatment with verteporfin, a YAP inhibitor, significantly reduced expression of fibrosis markers. In summary, these results suggest that c-Src has a critical role in cell fate determination during endodermal commitment of human iPSCs, and its alteration in early liver development in human may lead to increased production of abnormal YAP expressing profibrogenic proinflammatory cholangiocytes, similar to those seen in livers of patients with biliary fibrosis. Stem Cells 2019;37:306-317.


Asunto(s)
Proteína Tirosina Quinasa CSK/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Endodermo/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Conductos Biliares/enzimología , Conductos Biliares/patología , Proteína Tirosina Quinasa CSK/metabolismo , Endodermo/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Hígado/enzimología , Hígado/patología
3.
Stem Cell Res ; 24: 25-28, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29034890

RESUMEN

Biliary atresia (BA) is a common cause of pediatric end-stage liver disease. While its etiology is not yet clear, evidence has suggested that BA results from interactions between genetic susceptibility and environmental factors. Disease relevant human cellular models of BA will facilitate identification of both genetic and environmental factors that are important for disease prevention and treatment. Here we report the generation of a human induced pluripotent stem cell line from a BA patient using episomal vectors. Patient-specific BA iPSC lines provide valuable tools for disease mechanism study and drug development.


Asunto(s)
Atresia Biliar/etiología , Células Madre Pluripotentes Inducidas/metabolismo , Atresia Biliar/patología , Línea Celular , Preescolar , Humanos , Masculino
4.
Exp Biol Med (Maywood) ; 241(15): 1653-62, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27390263

RESUMEN

Due to the limitations of research using human embryos and the lack of a biological model of human liver development, the roles of the various markers associated with liver stem or progenitor cell potential in humans are largely speculative, and based on studies utilizing animal models and certain patient tissues. Human pluripotent stem cell-based in vitro multistage hepatic differentiation systems may serve as good surrogate models for mimicking normal human liver development, pathogenesis and injury/regeneration studies. Here, we describe the implications of various liver stem or progenitor cell markers and their bipotency (i.e. hepatocytic- and biliary-epithelial cell differentiation), based on the pluripotent stem cell-derived model of human liver development. Future studies using the human cellular model(s) of liver and biliary development will provide more human relevant biological and/or pathological roles of distinct markers expressed in heterogeneous liver stem/progenitor cell populations.


Asunto(s)
Sistema Biliar/crecimiento & desarrollo , Diferenciación Celular/fisiología , Hepatocitos/fisiología , Hígado/crecimiento & desarrollo , Células Madre/fisiología , Animales , Sistema Biliar/citología , Biomarcadores , Humanos , Hígado/citología
5.
Methods Mol Biol ; 1357: 383-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25410290

RESUMEN

The advent of induced pluripotent stem cell (iPSC) technology has enabled the modeling of an array of specific human disease phenotypes, aiding in the increasingly important and indispensable understanding of disease progression and pathogenesis. Pluripotent stem cell-derived hepatocytes present a new avenue for drug screening and personalized drug testing toward precision medicine. CYP450 microsomal enzymes play a critical role in drug metabolism. Hence, CYP activity measurement of iPSC-derived hepatocytes is a vital prerequisite, to ensure metabolic functionality before proceeding to drug testing. Herein, we describe the protocol for measurement of different CYP450 enzyme activities in human iPSC-derived hepatocytes.


Asunto(s)
Reprogramación Celular , Sistema Enzimático del Citocromo P-450/biosíntesis , Hepatocitos/enzimología , Células Madre Pluripotentes Inducidas/citología , Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Cultivadas , Técnicas de Reprogramación Celular/métodos , Medios de Cultivo/farmacología , Sistema Enzimático del Citocromo P-450/genética , Expresión Génica , Hepatocitos/citología , Humanos , Mediciones Luminiscentes , Microsomas Hepáticos/enzimología , ARN Mensajero/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
6.
Antivir Ther ; 20(8): 805-14, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26109548

RESUMEN

BACKGROUND: Lambda interferons (IFNLs) have potent antiviral activity against HCV, and polymorphisms within the IFNL gene cluster near the IFNL3 gene strongly predict spontaneous- and treatment-related HCV infection outcomes. The mechanism(s) linking IFNL polymorphisms and HCV control is currently elusive. METHODS: IFNL induction was studied in primary human hepatocytes (PHH) from 18 human donors, peripheral blood mononuclear cells (PBMCs) from 18 human donors, multiple cell lines and induced pluripotent stem cell-derived hepatocyte-like cells (iPSC-hepatocytes) from 7 human donors. After stimulation with intracellular RNA and infectious HCV, quantitative PCR (qPCR) primers and probes were designed to distinguish and quantify closely related IFNL messenger (m)RNAs from IFNL1, IFNL2 and IFNL3. RESULTS: PHH demonstrated the most potent induction of IFNLs, although had lower pre-stimulation levels compared to PBMCs, monocytes and cell lines. PHH stimulation with cytoplasmic poly I:C induced >1,000-fold expression of IFNL1, IFNL2 and IFNL3. PHH from donors who were homozygous for the favourable IFNL3 allele (IFNL3-CC) had higher IFNL3 induction compared to PHH from IFNL3-TT donors (P=0.03). Baseline IFNL mRNA expression and induction was also tested in iPSC-hepatocytes: iPSC-hepatocytes had significantly higher baseline expression of IFNLs compared to PHH (P<0.0001), and IFNL3 induction was marginally different in iPSC-hepatocytes by IFNL genotype (P=0.07). CONCLUSIONS: Hepatocytes express IFNLs when stimulated by a synthetic viral RNA that signals the cell through the cytoplasm. IFNL induction may be greater in persons with the favourable IFNL3 allele. These data provide insight into the strong linkage between IFNL3 genetics and control of HCV infection.


Asunto(s)
Regulación de la Expresión Génica , Genotipo , Hepatocitos/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Adolescente , Adulto , Anciano , Línea Celular , Niño , Femenino , Hepacivirus , Hepatitis C/genética , Hepatitis C/metabolismo , Hepatitis C/virología , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Interferones , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética , Adulto Joven
7.
Mol Ther ; 23(3): 570-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25418680

RESUMEN

Efficient and precise genome editing is crucial for realizing the full research and therapeutic potential of human induced pluripotent stem cells (iPSCs). Engineered nucleases including CRISPR/Cas9 and transcription activator like effector nucleases (TALENs) provide powerful tools for enhancing gene-targeting efficiency. In this study, we investigated the relative efficiencies of CRISPR/Cas9 and TALENs in human iPSC lines for inducing both homologous donor-based precise genome editing and nonhomologous end joining (NHEJ)-mediated gene disruption. Significantly higher frequencies of NHEJ-mediated insertions/deletions were detected at several endogenous loci using CRISPR/Cas9 than using TALENs, especially at nonexpressed targets in iPSCs. In contrast, comparable efficiencies of inducing homologous donor-based genome editing were observed at disease-associated loci in iPSCs. In addition, we investigated the specificity of guide RNAs used in the CRISPR/Cas9 system in targeting disease-associated point mutations in patient-specific iPSCs. Using myeloproliferative neoplasm patient-derived iPSCs that carry an acquired JAK2-V617F point mutation and α1-antitrypsin (AAT) deficiency patient-derived iPSCs that carry an inherited Z-AAT point mutation, we demonstrate that Cas9 can specifically target either the mutant or the wild-type allele with little disruption at the other allele differing by a single nucleotide. Overall, our results demonstrate the advantages of the CRISPR/Cas9 system in allele-specific genome targeting and in NHEJ-mediated gene disruption.


Asunto(s)
Alelos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genoma Humano , Células Madre Pluripotentes Inducidas/metabolismo , Sitios de Carácter Cuantitativo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Línea Celular , Reparación del ADN por Unión de Extremidades , Endonucleasas/genética , Endonucleasas/metabolismo , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Datos de Secuencia Molecular , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Reparación del ADN por Recombinación , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
8.
Antioxid Redox Signal ; 20(12): 1881-90, 2014 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-23066813

RESUMEN

SIGNIFICANCE: Stem cells are characterized by the properties of self-renewal and the ability to differentiate into multiple cell types, and thus maintain tissue homeostasis. Reactive oxygen species (ROS) are a natural byproduct of aerobic metabolism and have roles in cell signaling. Regulation of ROS has a vital role in maintaining "stemness" and differentiation of the stem cells, as well as in progression of stem-cell-associated diseases. RECENT ADVANCES: As of late, much research has been done on the adverse effects of ROS in stem cells. However, recently it has become apparent that in some cases redox status of the stem cell does have a role in maintaining its identity as such. Both pluripotent and multipotent stem cell types have been reported to possess enzymatic and nonenzymatic mechanisms for detoxification of ROS and to correct oxidative damage to the genome as well as the proteome. CRITICAL ISSUES: Although context dependent and somewhat varied among different stem cell types, the correlation seems to exist between antioxidant defense level and stem cell fate change (i.e., proliferation, differentiation, and death). Changes in stem cell redox regulation may affect the pathogenesis of various human diseases. FUTURE DIRECTIONS: Dissecting the defined roles of ROS in distinct stem cell types will greatly enhance their basic and translational applications. Here, we discuss the various roles of ROS in adult, embryonic, and induced pluripotent stem cells.


Asunto(s)
Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología , Hipoxia de la Célula , Linaje de la Célula , Humanos , Estrés Oxidativo , Células Madre/metabolismo
9.
Cell Cycle ; 10(15): 2423-7, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21750407

RESUMEN

Human induced pluripotent stem cells (iPSCs) are potential renewable sources of hepatocytes for drug development and cell therapy. Differentiation of human iPSCs into different developmental stages of hepatic cells has been achieved and improved during the last several years. We have recently demonstrated the liver engraftment and regenerative capabilities of human iPSC-derived multistage hepatic cells in vivo. Here we describe the in vitro and in vivo activities of hepatic cells derived from patient specific iPSCs, including multiple lines established from either inherited or acquired liver diseases, and discuss basic and clinical applications of these cells for disease modeling, drug screening and discovery, gene therapy and cell replacement therapy.


Asunto(s)
Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Evaluación de Medicamentos , Hepatocitos/trasplante , Humanos , Hepatopatías/prevención & control , Hepatopatías/terapia
10.
Blood ; 118(7): 1801-5, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21628406

RESUMEN

EBV-immortalized B lymphocyte cell lines have been widely banked for studying a variety of diseases, including rare genetic disorders. These cell lines represent an important resource for disease modeling with the induced pluripotent stem cell (iPSC) technology. Here we report the generation of iPSCs from EBV-immortalized B-cell lines derived from multiple inherited disease patients via a nonviral method. The reprogramming method for the EBV cell lines involves a distinct protocol compared with that of patient fibroblasts. The B-cell line-derived iPSCs expressed pluripotency markers, retained the inherited mutation and the parental V(D)J rearrangement profile, and differentiated into all 3 germ layer cell types. There was no integration of the reprogramming-related transgenes or the EBV-associated genes in these iPSCs. The ability to reprogram the widely banked patient B-cell lines will offer an unprecedented opportunity to generate human disease models and provide novel drug therapies.


Asunto(s)
Linfocitos B/citología , Linfocitos B/virología , Herpesvirus Humano 4/fisiología , Células Madre Pluripotentes Inducidas/citología , Linfocitos B/metabolismo , Diferenciación Celular , Línea Celular , Transformación Celular Viral , Células Cultivadas , Reprogramación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Herpesvirus Humano 4/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Transgenes
11.
Int J Biol Sci ; 6(7): 796-805, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21179587

RESUMEN

The recent advances in the induced pluripotent stem cell (iPSC) research have significantly changed our perspectives on regenerative medicine by providing researchers with a unique tool to derive disease-specific stem cells for study. In this review, we describe the human iPSC generation from developmentally diverse origins (i.e. endoderm-, mesoderm-, and ectoderm- tissue derived human iPSCs) and multistage hepatic differentiation protocols, and discuss both basic and clinical applications of these cells including disease modeling, drug toxicity screening/drug discovery, gene therapy and cell replacement therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Hepatopatías/patología , Tratamiento Basado en Trasplante de Células y Tejidos , Evaluación Preclínica de Medicamentos/métodos , Terapia Genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hepatopatías/tratamiento farmacológico , Hepatopatías/metabolismo , Hepatopatías/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...