Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Mol Biol Cell ; 30(24): 2969-2984, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31577526

RESUMEN

The apical surface of the terminally differentiated mammalian urothelial umbrella cell is mechanically stable and highly impermeable, in part due to its coverage by urothelial plaques consisting of 2D crystals of uroplakin particles. The mechanism for regulating the uroplakin/plaque level is unclear. We found that genetic ablation of the highly tissue-specific sorting nexin Snx31, which localizes to plaques lining the multivesicular bodies (MVBs) in urothelial umbrella cells, abolishes MVBs suggesting that Snx31 plays a role in stabilizing the MVB-associated plaques by allowing them to achieve a greater curvature. Strikingly, Snx31 ablation also induces a massive accumulation of uroplakin-containing mitochondria-derived lipid droplets (LDs), which mediate uroplakin degradation via autophagy/lipophagy, leading to the loss of apical and fusiform vesicle plaques. These results suggest that MVBs play an active role in suppressing the excessive/wasteful endocytic degradation of uroplakins. Failure of this suppression mechanism triggers the formation of mitochondrial LDs so that excessive uroplakin membranes can be sequestered and degraded. Because mitochondrial LD formation, which occurs at a low level in normal urothelium, can also be induced by disturbance in uroplakin polymerization due to individual uroplakin knockout and by arsenite, a bladder carcinogen, this pathway may represent an inducible, versatile urothelial detoxification mechanism.


Asunto(s)
Cuerpos Multivesiculares/metabolismo , Nexinas de Clasificación/metabolismo , Urotelio/metabolismo , Animales , Femenino , Gotas Lipídicas/metabolismo , Gotas Lipídicas/fisiología , Glicoproteínas de Membrana/metabolismo , Membranas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Vejiga Urinaria/metabolismo , Uroplaquinas/metabolismo , Uroplaquinas/fisiología
2.
PLoS One ; 9(6): e99644, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24914955

RESUMEN

Uroplakins (UP), a group of integral membrane proteins, are major urothelial differentiation products that form 2D crystals of 16-nm particles (urothelial plaques) covering the apical surface of mammalian bladder urothelium. They contribute to the urothelial barrier function and, one of them, UPIa, serves as the receptor for uropathogenic Escherichia coli. It is therefore important to understand the mechanism by which these surface-associated uroplakins are degraded. While it is known that endocytosed uroplakin plaques are targeted to and line the multivesicular bodies (MVBs), it is unclear how these rigid-looking plaques can go to the highly curved membranes of intraluminal vesicles (ILVs). From a cDNA subtraction library, we identified a highly urothelium-specific sorting nexin, SNX31. SNX31 is expressed, like uroplakins, in terminally differentiated urothelial umbrella cells where it is predominantly associated with MVBs. Apical membrane proteins including uroplakins that are surface biotin-tagged are endocytosed and targeted to the SNX31-positive MVBs. EM localization demonstrated that SNX31 and uroplakins are both associated not only with the limiting membranes of MVBs containing uroplakin plaques, but also with ILVs. SNX31 can bind, on one hand, the PtdIns3P-enriched lipids via its N-terminal PX-domain, and, on the other hand, it binds uroplakins as demonstrated by co-immunoprecipitation and proximity ligation assay, and by its reduced membrane association in uroplakin II-deficient urothelium. The fact that in urothelial umbrella cells MVBs are the only major intracellular organelles enriched in both PtdIns3P and uroplakins may explain SNX31's MVB-specificity in these cells. However, in MDCK and other cultured cells transfected SNX31 can bind to early endosomes possibly via lipids. These data support a model in which SNX31 mediates the endocytic degradation of uroplakins by disassembling/collapsing the MVB-associated uroplakin plaques, thus enabling the uroplakin-containing (but 'softened') membranes to bud and form the ILVs for lysosomal degradation and/or exosome formation.


Asunto(s)
Diferenciación Celular , Cuerpos Multivesiculares/metabolismo , Nexinas de Clasificación/metabolismo , Uroplaquinas/metabolismo , Urotelio/citología , Urotelio/metabolismo , Animales , Biomarcadores/metabolismo , Bovinos , Membrana Celular/metabolismo , Perros , Endocitosis , Endosomas/metabolismo , Técnicas de Inactivación de Genes , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Modelos Biológicos , Cuerpos Multivesiculares/ultraestructura , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica , Ultracentrifugación , Urotelio/enzimología , Urotelio/ultraestructura
3.
Cell Res ; 22(1): 194-207, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21862970

RESUMEN

Embryonic hematopoiesis is a complex process. Elucidating the mechanism regulating hematopoietic differentiation from pluripotent stem cells would allow us to establish a strategy to efficiently generate hematopoietic cells. However, the mechanism governing the generation of hematopoietic progenitors from human embryonic stem cells (hESCs) remains unknown. Here, on the basis of the emergence of CD43(+) hematopoietic cells from hemogenic endothelial (HE) cells, we demonstrated that VEGF was essential and sufficient, and that bFGF was synergistic with VEGF to specify the HE cells and the subsequent transition into CD43(+) hematopoietic cells. Significantly, we identified TGFß as a novel signal to regulate hematopoietic development, as the TGFß inhibitor SB 431542 significantly promoted the transition from HE cells into CD43(+) hematopoietic progenitor cells (HPCs) during hESC differentiation. By defining these critical signaling factors during hematopoietic differentiation, we can efficiently generate HPCs from hESCs. Our strategy could offer an in vitro model to study early human hematopoietic development.


Asunto(s)
Hemangioblastos/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Activinas/farmacología , Animales , Benzamidas/farmacología , Diferenciación Celular , Línea Celular , Proliferación Celular , Dioxoles/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Factores de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Hemangioblastos/citología , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucosialina/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
PLoS One ; 6(8): e23383, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21826251

RESUMEN

BACKGROUND: Pancreatic cancer is one of the direct causes of cancer-related death. High level of chemoresistance is one of the major obstacles of clinical treatment. In recent years, cancer stem cells have been widely identified and indicated as the origin of chemoresistance in multi-types of solid tumors. Increasing evidences suggest that cancer stem cells reside in the cells capable of forming holoclones continuously. However, in pancreatic cancer, holoclone-forming cells have not been characterized yet. Therefore, the goal of our present study was to indentify the holoclone-forming pancreatic cancer stem cells and develop an in vitro continuous colony formation system, which will greatly facilitate the study of pancreatic cancer stem cells. METHODOLOGY/PRINCIPAL FINDINGS: Pancreatic cancer cell line BxPC3 was submitted to monoclonal cultivation to generate colonies. Based on the morphologies, colonies were classified and analyzed for their capacities of secondary colony formation, long-term survival in vitro, tumor formation in vivo, and drug resistance. Flowcytometry and quantitative RT-PCR were performed to detect the expression level of cancer stem cells associated cell surface markers, regulatory genes and microRNAs in distinct types of colonies. Three types of colonies with distinct morphologies were identified and termed as holo-, mero-, and paraclones, in which only holoclones generated descendant colonies of all three types in further passages. Compared to mero- and paraclones, holoclones possessed higher capacities of long-term survival, tumor initiation, and chemoresistance. The preferential expression of cancer stem cells related marker (CXCR4), regulatory genes (BMI1, GLI1, and GLI2) and microRNAs (miR-214, miR-21, miR-221, miR-222 and miR-155) in holoclones were also highlighted. CONCLUSIONS/SIGNIFICANCE: Our results indicate that the pancreatic tumor-initiating cells with high level of chemoresistance were enriched in holoclones derived from BxPC3 cell line. Generation of holoclones can serve as a novel model for studying cancer stem cells, and attribute to developing new anti-cancer drugs.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Citometría de Flujo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/metabolismo , Receptores CXCR4/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
5.
Stem Cells ; 29(4): 609-17, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21308865

RESUMEN

Human ESCs provide a promising cell resource for the treatment of type I diabetes mellitus. Although PDX1-positive pancreatic progenitors can be efficiently generated from human ESCs by stepwise induction, further in vitro differentiation into functional, mature beta cells is not efficient or reproducible. Purification of pancreatic progenitor cells could facilitate the identification of signals that regulate beta cell differentiation and maturation. Here, we report the identification of a novel surface marker for PDX1-positive pancreatic progenitors based on an in vitro human ESC differentiation system. By costaining PDX1 and a panel of cell surface antigens at the pancreatic progenitor stage of human ESC differentiation, we discovered a positive marker, CD24. CD24-positive cells coexpressed most of the key transcription factors of pancreatic progenitors, and the expression of important pancreatic genes was greatly enriched in CD24-positive cells compared with the CD24-negative cells. In addition, CD24-positive cells could differentiate into insulin-producing cells but CD24-negative cells could not. These results indicate that CD24 could be a surface marker for PDX1-positive pancreatic progenitors derived from human ESCs. Enrichment of pancreatic progenitors with this marker will facilitate the investigation of beta cell maturation during the human ESC differentiation.


Asunto(s)
Antígeno CD24/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Secretoras de Insulina/inmunología , Páncreas/citología , Antígenos de Superficie/inmunología , Biomarcadores , Antígeno CD24/inmunología , Diferenciación Celular , Línea Celular , Diabetes Mellitus Tipo 1/terapia , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/biosíntesis , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/biosíntesis
6.
Blood ; 116(23): 4786-94, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20427702

RESUMEN

Current induction schemes directing hematopoietic differentiation of human embryonic stem cells (hESCs) are not well defined to mimic the sequential stages of hematopoietic development in vivo. Here, we report a 3-stage method to direct differentiation of hESCs toward hematopoietic progenitors in chemically defined mediums. In the first 2 stages, we efficiently generated T-positive primitive streak/mesendoderm cells and kinase domain receptor-positive (KDR(+)) platelet-derived growth factor receptor α-negative (PDGFRα(-)) hemato-vascular precursors sequentially. In the third stage, we found that cells in a spontaneous differentiation condition mainly formed erythroid colonies. Addition of all-trans retinoic acid (RA) greatly enhanced generation of hematopoietic progenitors in this stage while suppressing erythroid development. The RA-treated cells highly expressed definitive hematopoietic genes, formed large numbers of multilineage and myeloid colonies, and gave rise to greater than 45% CD45(+) hematopoietic cells. When hematopoietic progenitors were selected with CD34 and C-Kit, greater than 95% CD45(+) hematopoietic cells could be generated. In addition, we found that endogenous RA signaling at the second stage was required for vascular endothelial growth factor/basic fibroblast growth factor-induced hemato-vascular specification, whereas exogenously applied RA efficiently induced KDR(-)PDGFRα(+) paraxial mesoderm cells. Our study suggests that RA signaling plays diverse roles in human mesoderm and hematopoietic development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Tretinoina/metabolismo , Diferenciación Celular/efectos de los fármacos , Separación Celular , Células Madre Embrionarias/efectos de los fármacos , Citometría de Flujo , Hematopoyesis/efectos de los fármacos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tretinoina/farmacología
7.
J Mol Cell Biol ; 2(1): 50-60, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19910415

RESUMEN

One key step in producing insulin-secreting cells from human embryonic stem (hES) cells is the generation of pancreatic and duodenal homeobox gene 1 (PDX1)-expressing pancreatic progenitor cells. All-trans retinoic acid (RA) has important roles in pancreas development and is widely used to induce pancreatic differentiation of ES cells. When RA was added directly to the activin A-induced hES cells, <20% cells were positive for the pancreatic marker PDX1, whereas the other cells were mainly hepatic cells. We found that when the activin A-induced hES cells were replated and seeded at low cell densities, the addition of RA induced significant pancreatic differentiation and over 70% of cells in culture expressed PDX1. When the endodermal cells were isolated with the surface marker CXCR4 from the activin A-induced culture and further differentiated with RA, a homogeneous PDX1(+) cell population (over 95% pure) was generated. The PDX1(+) cells could further differentiate into cells that expressed pancreatic transcription factors and pancreatic endocrine or exocrine markers. We also found that RA inhibited the hepatic differentiation of endodermal cells that were seeded at low cell densities, and this inhibition may have been through the inhibition of Smad1/5/8 activity. Thus, we present a highly efficient and reproducible protocol for generating PDX1(+) pancreatic progenitor cells from hES cells.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Células Madre Embrionarias/citología , Endodermo/citología , Proteínas de Homeodominio/metabolismo , Páncreas/metabolismo , Transactivadores/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Proteínas de Homeodominio/genética , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Páncreas/citología , Transactivadores/genética , Tretinoina/metabolismo
8.
Dev Dyn ; 239(2): 407-24, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20014101

RESUMEN

Maternal effect genes and encoding proteins are necessary for nuclear reprogramming and zygotic genome activation. However, the mechanisms that mediate these functions are largely unknown. Here we identified XM_359149, a Zar1-like gene that is predominantly expressed in oocytes and zygotes, which we designated Zar1-like (Zar1l). ZAR1L-EGFP formed multiple cytoplasmic foci in late two-cell-stage embryos. Expression of the ZAR1L C-terminus induced two-cell-stage embryonic arrest, accompanied with abnormal methylation of histone H3K4me2/3 and H3K9me2/3, and marked down-regulation of a group of chromatin modification factors including Dppa2, Dppa4, and Piwil2. When ectopically expressed in somatic cells, ZAR1L colocalized with P-body components including EIF2C1(AGO1), EIF2C2(AGO2), DDX6 and LSM14A, and germline-specific chromatoid body components including PIWIL1, PIWIL2, and LIN28. ZAR1L colocalized with ZAR1 and interacted with human LIN28. Our data suggest that ZAR1L and ZAR1 may comprise a novel family of processing-body/chromatoid-body components that potentially function as RNA regulators in early embryos.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Argonautas , Bromodesoxiuridina , Línea Celular , ARN Helicasas DEAD-box/metabolismo , Proteínas del Huevo/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Mutación , Proteínas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Alineación de Secuencia , Factores de Transcripción
9.
PLoS One ; 4(12): e8344, 2009 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20020058

RESUMEN

Under normal conditions, the regeneration of mouse beta cells is mainly dependent on their own duplication. Although there is evidence that pancreatic progenitor cells exist around duct, whether non-beta cells in the islet could also potentially contribute to beta cell regeneration in vivo is still controversial. Here, we developed a novel transgenic mouse model to study the pancreatic beta cell regeneration, which could specifically inhibit beta cell proliferation by overexpressing p21(cip) in beta cells via regulation of the Tet-on system. We discovered that p21 overexpression could inhibit beta-cell duplication in the transgenic mice and these mice would gradually suffer from hyperglycemia. Importantly, the recovery efficiency of the p21-overexpressing mice from streptozotocin-induced diabetes was significantly higher than control mice, which is embodied by better physiological quality and earlier emergence of insulin expressing cells. Furthermore, in the islets of these streptozotocin-treated transgenic mice, we found a large population of proliferating cells which expressed pancreatic duodenal homeobox 1 (PDX1) but not markers of terminally differentiated cells. Transcription factors characteristic of early pancreatic development, such as Nkx2.2 and NeuroD1, and pancreatic progenitor markers, such as Ngn3 and c-Met, could also be detected in these islets. Thus, our work showed for the first time that when beta cell self-duplication is repressed by p21 overexpression, the markers for embryonic pancreatic progenitor cells could be detected in islets, which might contribute to the recovery of these transgenic mice from streptozotocin-induced diabetes. These discoveries could be important for exploring new diabetes therapies that directly promote the regeneration of pancreatic progenitors to differentiate into islet beta cells in vivo.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Hormonas/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/efectos de los fármacos , Transactivadores/metabolismo
10.
Am J Pathol ; 175(5): 1975-83, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19808649

RESUMEN

Mouse models of liver injury provide useful tools for studying hepatocyte engraftment and proliferation. A representative model of liver injury is the albumin-urokinase (Alb-uPA) transgenic model, but neonatal lethality hampers its widespread application. To overcome this problem, we generated a transgenic mouse in which transcription of the reverse tetracycline transactivator was (rtTA) driven by the mouse albumin promoter, and backcrossed the rtTA mice onto severe combined immunodeficient (SCID)/bg mice to generate immunodeficient rtTA/SCID mice. We then produced recombinant adenoviruses Ad.TRE-uPA, in which the urokinase was located downstream of the tetracycline response element (TRE). The rtTA/SCID mouse hepatocytes were then infected with Ad.TRE-uPA to establish an inducible liver injury mouse model. In the presence of doxycycline, uPA was exclusively expressed in endogenous hepatocytes and caused extensive liver injury. Enhanced green fluorescent protein-labeled mouse hepatocytes selectively repopulated the rtTA/SCID mouse liver and replaced over 80% of the recipient liver mass after repeated administration of Ad.TRE-uPA. Compared with the original uPA mice, rtTA/SCID mice did not exhibit problems regarding breeding efficiency, and the time window for transplantation was flexible. In addition, we could control the extent of liver injury to facilitate transplantation surgery by regulating the dose of Ad.TRE-uPA. Our inducible mouse model will be convenient for studies of hepatocyte transplantation and hepatic regeneration, and this system will facilitate screening for potential genetic factors critical for engraftment and proliferation of hepatocytes in vivo.


Asunto(s)
Hepatocitos/trasplante , Hígado , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Albúminas/genética , Albúminas/metabolismo , Animales , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/citología , Hepatocitos/fisiología , Hígado/citología , Hígado/enzimología , Hígado/lesiones , Trasplante de Hígado , Ratones , Ratones SCID , Ratones Transgénicos , Modelos Animales , Tetraciclina/metabolismo , Transaminasas/sangre , Activador de Plasminógeno de Tipo Uroquinasa/genética
11.
Mol Cell ; 35(6): 841-55, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19782033

RESUMEN

Cul3, a Cullin family scaffold protein, is thought to mediate the assembly of a large number of SCF (Skp1-Cullin1-F-box protein)-like ubiquitin ligase complexes through BTB domain substrate-recruiting adaptors. Cul3 controls early embryonic development in several genetic models through mechanisms not understood. Very few functional substrate/adaptor pairs for Cul3 ubiquitin ligases have been identified. Here, we show that Cul3 knockdown in human cells results in abnormal actin stress fibers and distorted cell morphology, owing to impaired ubiquitination and degradation of small GTPase RhoA. We identify a family of RhoA-binding BTB domain adaptors conserved from insects to mammals, designated BACURDs. BACURDs form ubiquitin ligase complexes, which selectively ubiquitinate RhoA, with Cul3. Dysfunction of the Cul3/BACURD complex decreases cell migration potential and impairs RhoA-mediated convergent extension movements during Xenopus gastrulation. Our studies reveal a previously unknown mechanism for controlling RhoA degradation and regulating RhoA function in various biological contexts, which involves a Cul3/BACURD ubiquitin ligase complex.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Proteínas Cullin/metabolismo , Proteínas de Drosophila/metabolismo , Procesamiento Proteico-Postraduccional , Fibras de Estrés/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Forma de la Célula , Secuencia Conservada , Proteínas Cullin/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Evolución Molecular , Gastrulación , Células HeLa , Humanos , Complejos Multiproteicos , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Transfección , Ubiquitinación , Xenopus/embriología , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
12.
Cell Res ; 19(11): 1233-42, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19736565

RESUMEN

Human induced pluripotent stem (iPS) cells are similar to embryonic stem (ES) cells, and can proliferate intensively and differentiate into a variety of cell types. However, the hepatic differentiation of human iPS cells has not yet been reported. In this report, human iPS cells were induced to differentiate into hepatic cells by a stepwise protocol. The expression of liver cell markers and liver-related functions of the human iPS cell-derived cells were monitored and compared with that of differentiated human ES cells and primary human hepatocytes. Approximately 60% of the differentiated human iPS cells at day 7 expressed hepatic markers alpha fetoprotein and Alb. The differentiated cells at day 21 exhibited liver cell functions including albumin Asecretion, glycogen synthesis, urea production and inducible cytochrome P450 activity. The expression of hepatic markers and liver-related functions of the iPS cell-derived hepatic cells were comparable to that of the human ES cell-derived hepatic cells. These results show that human iPS cells, which are similar to human ES cells, can be efficiently induced to differentiate into hepatocyte-like cells.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Hepatocitos/citología , Células Madre Pluripotentes Inducidas , Línea Celular , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Células Madre Embrionarias/metabolismo , Glicoproteínas/metabolismo , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Glucógeno Hepático/biosíntesis , Albúmina Sérica/metabolismo , Albúmina Sérica Humana , Urea/metabolismo , alfa-Fetoproteínas/biosíntesis
13.
PLoS One ; 4(7): e6468, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19649295

RESUMEN

The derivation of hepatic progenitor cells from human embryonic stem (hES) cells is of value both in the study of early human liver organogenesis and in the creation of an unlimited source of donor cells for hepatocyte transplantation therapy. Here, we report for the first time the generation of hepatic progenitor cells derived from hES cells. Hepatic endoderm cells were generated by activating FGF and BMP pathways and were then purified by fluorescence activated cell sorting using a newly identified surface marker, N-cadherin. After co-culture with STO feeder cells, these purified hepatic endoderm cells yielded hepatic progenitor colonies, which possessed the proliferation potential to be cultured for an extended period of more than 100 days. With extensive expansion, they co-expressed the hepatic marker AFP and the biliary lineage marker KRT7 and maintained bipotential differentiation capacity. They were able to differentiate into hepatocyte-like cells, which expressed ALB and AAT, and into cholangiocyte-like cells, which formed duct-like cyst structures, expressed KRT19 and KRT7, and acquired epithelial polarity. In conclusion, this is the first report of the generation of proliferative and bipotential hepatic progenitor cells from hES cells. These hES cell-derived hepatic progenitor cells could be effectively used as an in vitro model for studying the mechanisms of hepatic stem/progenitor cell origin, self-renewal and differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Hígado/citología , Cadherinas/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Cell Res ; 19(6): 710-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19365404

RESUMEN

PTEN, a negative regulator of the phosphatidylinositol-3-kinase/AKT pathway, is an important modulator of insulin signaling. To determine the metabolic function of pancreatic Pten, we generated pancreas-specific Pten knockout (PPKO) mice. PPKO mice had enlarged pancreas and elevated proliferation of acinar cells. They also exhibited hypoglycemia, hypoinsulinemia, and altered amino metabolism. Notably, PPKO mice showed delayed onset of streptozotocin (STZ)-induced diabetes and sex-biased resistance to high-fat-diet (HFD)-induced diabetes. To investigate the mechanism for the resistance to HFD-induced hyperglycemia in PPKO mice, we evaluated AKT phosphorylation in major insulin-responsive tissues: the liver, muscle, and fat. We found that Pten loss in the pancreas causes the elevation of AKT signaling in the liver. The phosphorylation of AKT and its downstream substrate GSK3beta was increased in the liver of PPKO mice, while PTEN level was decreased without detectable excision of Pten allele in the liver of PPKO mice. Proteomics analysis revealed dramatically decreased level of 78-kDa glucose-regulated protein (GRP78) in the liver of PPKO mice, which may also contribute to the lower blood glucose level of PPKO mice fed with HFD. Together, our findings reveal a novel response in the liver to pancreatic defect in metabolic regulation, adding a new dimension to understanding diabetes resistance.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Hígado/enzimología , Fosfohidrolasa PTEN/metabolismo , Páncreas/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proliferación Celular , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Grasas de la Dieta/farmacología , Chaperón BiP del Retículo Endoplásmico , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Choque Térmico/metabolismo , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosforilación , Proteómica , Transducción de Señal
15.
Cell Stem Cell ; 3(6): 587-90, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19041774

RESUMEN

Induced pluripotent stem (iPS) cells can be generated from somatic cells by transduction with several transcription factors in mouse and human. However, direct reprogramming in other species has not been reported. Here, we generated monkey iPS cells by retrovirus-mediated introduction of monkey transcription factors OCT4, SOX2, KLF4, and c-MYC.


Asunto(s)
Diferenciación Celular/genética , Fibroblastos/metabolismo , Macaca mulatta/fisiología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/genética , Transducción Genética/métodos , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Forma de la Célula/genética , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Desarrollo Embrionario/genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Macaca mulatta/anatomía & histología , Masculino , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Proteínas Proto-Oncogénicas c-myc/genética , Retroviridae/genética , Factores de Transcripción SOXB1/genética
17.
Differentiation ; 76(9): 923-30, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18557766

RESUMEN

Feeder cells are commonly used to culture embryonic stem cells to maintain their undifferentiated and pluripotent status. Conventionally, mouse embryonic fibroblasts (MEFs), supplemented with leukemia inhibitory factor (LIF), are used as feeder cells to support the growth of mouse embryonic stem cells (mESCs) in culture. To prepare for fresh MEF feeder or for MEF-conditioned medium, sacrifice of mouse fetuses repeatedly is unavoidable in these tedious culture systems. Here we report the discovery of a human endothelial cell line (ECV-304 cell line) that efficiently supports growth of mESCs LIF-free conditions. mESCs that were successfully cultured for eight to 20 passages on ECV-304 feeders showed morphological characteristics similar to cells cultured in traditional feeder cell systems. These cells expressed the stem cell markers Oct3/4, Nanog, Sox2, and SSEA-1. Furthermore, cells cultured on the ECV-304 cell line were able to differentiate into three germ layers and were able to generate chimeric mice. Compared with traditional culture systems, there is no requirement for mouse fetuses and exogenous LIF does not need to be added to the culture system. As a stable cell line, the ECV-304 cell line efficiently replaces MEFs as an effective feeder system and allows the efficient expansion of mESCs.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Endoteliales/citología , Animales , Biomarcadores/metabolismo , Línea Celular , Técnicas de Cocultivo , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/farmacología , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
18.
Hum Reprod ; 23(1): 54-61, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18000170

RESUMEN

BACKGROUND: Mouse fetal germ cells have been successfully purified from fetal gonads. However, there are no published reports describing a procedure for deriving mature oocytes from isolated fetal germ cells. The purpose of this present study is to explore whether purified fetal germ cells are able to differentiate into mature oocytes through an in vivo grafting procedure. METHODS AND RESULTS: First, intact 11.5 and 12.5 days post-coitum (dpc) female gonads with or without the attached mesonephros and the reaggregated female gonad cells were transplanted into the recipient mice. The results demonstrate both the gonad accompanied by mesonephroi and the innate gonad structure are not absolutely required for 11.5 dpc and 12.5 dpc oogonia to generate mature oocytes. Next, oogonia were purified from female gonads, aggregated with different ovarian somatic cells and transplanted into the recipient mice. Purified 12.5 dpc oogonia were able to generate mature oocytes by aggregating with 12.5 dpc ovarian somatic cells, but not with 16.5 dpc or 0 days postpartum ovarian somatic cells. We also tested 12.5 dpc male germ cells but they were unable to undergo oogenesis. CONCLUSIONS: Our study demonstrates that mature oocytes can be derived from purified fetal germ cells through an aggregation and transplantation procedure. It also suggests that the synchronized interactions between oogonia and gonadal somatic cells are important to ensure normal folliculogenesis.


Asunto(s)
Embrión de Mamíferos/citología , Ratones/embriología , Oocitos/fisiología , Ovario/embriología , Animales , Agregación Celular , Diferenciación Celular , Femenino , Masculino , Mesonefro/trasplante , Ratones Endogámicos ICR , Oogonios/citología , Oogonios/trasplante , Ovario/trasplante , Trasplante Heterotópico
19.
Blood ; 111(4): 1933-41, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18042803

RESUMEN

Human embryonic stem cells (hES cells) have unlimited self-renewal capacity and can differentiate into most, if not all, possible cell types. This unique property makes them valuable not only for investigation of early developmental processes, but also for regenerative medicine. Mesoderm-derived cardiac cells and hematopoietic cells both have the potential for various therapeutic applications. However, efficient induction of hES cell differentiation into mesoderm remains a challenge. Here, we showed that treatment of hES cells with bone morphogenetic protein-4 (BMP-4) exhibited differential effects: long-term treatment results in trophoblast and extra-embryonic endoderm differentiation, whereas short-term treatment can promote early mesoderm induction. The induction of mesoderm in hES cells occurs at a high efficiency as measured using several markers, such as Brachyury, WNT3, and MIXL1 expression. Moreover, these mesoderm progenitor cells can differentiate into cardiac and hematopoietic lineages in vitro. Further analysis showed that the mesoderm-inducing capacity of BMP-4 requires endogenous FGF and TGF-beta/Nodal/activin signaling activities. Thus, our results uncover a novel role for BMP-4 in regulation of hES cell differentiation and should provide insights into the mechanism of mesoderm induction in hES cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Mesodermo/citología , Fosfatasa Alcalina/análisis , Animales , Biomarcadores/análisis , Proteína Morfogenética Ósea 4 , Técnicas de Cultivo de Célula , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Mesodermo/efectos de los fármacos , Mesodermo/fisiología , Ratones , Mitomicina/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Comp Med ; 57(5): 450-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17974127

RESUMEN

To establish a small animal model of severe acute respiratory syndrome (SARS), we developed a mouse model of human severe acute respiratory syndrome coronavirus (SARS-CoV) infection by introducing the human gene for angiotensin-converting enzyme 2 (hACE2) (the cellular receptor of SARS-CoV), driven by the mouse ACE2 promoter, into the mouse genome. The hACE2 gene was expressed in lung, heart, kidney, and intestine. We also evaluated the responses of wild-type and transgenic mice to SARS-CoV inoculation. At days 3 and 7 postinoculation, SARS-CoV replicated more efficiently in the lungs of transgenic mice than in those of wild-type mice. In addition, transgenic mice had more severe pulmonary lesions, including interstitial hyperemia and hemorrhage, monocytic and lymphocytic infiltration, protein exudation, and alveolar epithelial cell proliferation and desquamation. Other pathologic changes, including vasculitis, degeneration, and necrosis, were found in the extrapulmonary organs of transgenic mice, and viral antigen was found in brain. Therefore, transgenic mice were more susceptible to SARS-CoV than were wild-type mice, and susceptibility was associated with severe pathologic changes that resembled human SARS infection. These mice will be valuable for testing potential vaccine and antiviral drug therapies and for furthering our understanding of SARS pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Peptidil-Dipeptidasa A/genética , Síndrome Respiratorio Agudo Grave/virología , Enzima Convertidora de Angiotensina 2 , Animales , Femenino , Ingeniería Genética , Predisposición Genética a la Enfermedad , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos ICR , Peptidil-Dipeptidasa A/metabolismo , ARN Mensajero/metabolismo , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...