Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Sci ; 124(Pt 11): 1819-30, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21540297

RESUMEN

VEGF-, phosphoinositide 3-kinase (PI3K)- and protein kinase C (PKC)-regulated signaling in cardiac and vascular differentiation was investigated in mouse ES cells and in ES cell-derived Flk-1⁺ cardiovascular progenitor cells. Inhibition of PI3K by wortmannin and LY294002, disruption of PI3K catalytic subunits p110α and p110δ using short hairpin RNA (shRNA), or inhibition of p110α with compound 15e and of p110δ with IC-87114 impaired cardiac and vascular differentiation. By contrast, TGX-221, an inhibitor of p110ß, and shRNA knockdown of p110ß were without significant effects. Antagonists of the PKC family, i.e. bisindolylmaleimide-1 (BIM-1), GÖ 6976 (targeting PKCα/ßII) and rottlerin (targeting PKCδ) abolished vasculogenesis, but not cardiomyogenesis. Inhibition of Akt blunted cardiac as well as vascular differentiation. VEGF induced phosphorylation of PKCα/ßII and PKCδ but not PKCζ. This was abolished by PI3K inhibitors and the VEGFR-2 antagonist SU5614. Furthermore, phosphorylation of Akt and phosphoinositide-dependent kinase-1 (PDK1) was blunted upon inhibition of PI3K, but not upon inhibition of PKC by BIM-1, suggesting that activation of Akt and PDK1 by VEGF required PI3K but not PKC. In summary, we demonstrate that PI3K catalytic subunits p110α and p110δ are central to cardiovasculogenesis of ES cells. Akt downstream of PI3K is involved in both cardiomyogenesis and vasculogenesis, whereas PKC is involved only in vasculogenesis.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Cuerpos Embrioides/citología , Desarrollo de Músculos , Miocardio/citología , Neovascularización Fisiológica , Proteína Quinasa C/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Dominio Catalítico , Diferenciación Celular , Línea Celular , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Cuerpos Embrioides/efectos de los fármacos , Activación Enzimática , Ratones , Morfolinas/farmacología , Miocardio/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal
3.
Methods Mol Biol ; 331: 313-28, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16881524

RESUMEN

Human embryonic stem cells efficiently differentiate blood vessels, which allows using this in vitro model to study the interaction of blood vessels with adjacent tissues. Herein, we introduce confrontation cultures of human embryonic stem cells with multicellular tumor spheroids to investigate molecular mechanisms of tumor-induced angiogenesis. Vascularization of tumor tissue by the host is a prerequisite for tumor growth, which has led to the development of antiangiogenic therapy. This promising anti-cancer therapy intends to reduce, halt, or even regress tumor growth by deprivation from blood, oxygen, and nutrient supply. Confrontation cultures of human embryonic stem cells with multicellular tumor spheroids allow the investigation of the time course of endothelial cell invasion into the tumor tissue, the concomitant analysis of changes in angiogenesis-related gene expression, and analysis of the cellular microenvironment (i.e., pericellular oxygen pressure, tissue pH, and levels of tissue reactive oxygen species). The in vitro model of confrontation cultures is suitable for routine screening of antiangiogenic agents in pre-clinical trials and may be used to replace animal experiments applied in antiangiogenesis research.


Asunto(s)
Técnicas de Cocultivo/métodos , Neovascularización Patológica/patología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Esferoides Celulares/citología , Alternativas al Uso de Animales , Animales , Capilares/citología , Capilares/fisiología , Comunicación Celular , Células Endoteliales/citología , Humanos , Inmunohistoquímica , Microscopía Confocal , Células Tumorales Cultivadas/citología
5.
Reprod Toxicol ; 65: 46-58, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27346840

RESUMEN

Man-made magnetic fields (MFs) may exert adverse effects on mammalian embryonic development. Herein, we analysed the effect of 10mT 50Hz sinusoidal (AC) or static (DC) MFs versus 1mT MFs on embryonic development of mice. Exposure for 20days during gestation to 10mT MFs increased resorptions and dead fetuses, decreased crown-rump length and fresh weight, reduced blood vessel differentiation and caused histological changes, accompanied with diminished vascular endothelial growth factor (VEGF) protein expression in several organs. In embryonic stem (ES) cell-derived embryoid bodies exposure towards 10mT MFs increased reactive oxygen species (ROS), decreased vascular marker as well as VEGF expression and enhanced apoptosis. In conclusion, our combined data from in vivo and in vitro experiments identified VEGF as an important mediator during embryonic development that can be influenced by high strength MFs, which in consequence leads to severe abnormalities in fetus organs and blood vessel formation.


Asunto(s)
Desarrollo Embrionario , Campos Magnéticos , Neovascularización Fisiológica , Animales , Línea Celular , Largo Cráneo-Cadera , Pérdida del Embrión , Embrión de Mamíferos , Cuerpos Embrioides/metabolismo , Ojo/embriología , Ojo/metabolismo , Femenino , Hígado/embriología , Hígado/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Células Madre Embrionarias de Ratones/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Aumento de Peso
6.
Cytometry B Clin Cytom ; 86(4): 229-35, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23576291

RESUMEN

BACKGROUND: Paradoxical activation of the MAP-kinases, ERK1, and ERK2 (ERK1/2) is observed in CML cell lines and primary CML patient cells treated with tyrosine kinase inhibitors (TKI) in vitro. The commonly accepted assumption is that activated ERK1/2 is key regulators of survival of leukemic cells treated with kinase inhibitors. Hence, paradoxical ERK1/2-activation may trigger resistance in vivo, which yet has to be shown. We therefore sought to establish a flow cytometric assay that enables us to measure paradoxical TKI-induced ERK1/2-activation on a single cell basis in primary CML cells. METHODS: Side-by-side Western blot and intracellular flow cytometry (FCM) after in vitro exposure of cell lines and primary cells to nilotinib were performed. Detailed analysis of pre-analytical factors and the issue of compartmentalization of phosphorylated ERK1/2 by confocal laser scanning microscopy were performed. RESULT: Results were conflicting in that pERK-activation was robustly detected in Western blot assays, but not when cells were analyzed by FCM despite well functioning positive and negative controls. This is in contrast to experiments on other targets such as phospho-CrkL, where also in our hands TKI-dependent inhibition of phosphorylation is trackable by both Western blot and FCM assays. CONCLUSIONS: To our knowledge this is the first report of discordant results in phospho-protein analysis in TKI-treated cells analyzed by Western blot vs. FCM. We speculate that a substance specific interaction interferes with fluorescence dependent methods seeking to track phosphorylated ERK1/2 in TKI-treated cells.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antígenos CD34/metabolismo , Línea Celular Transformada , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
7.
Stem Cells Dev ; 23(4): 333-51, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24134622

RESUMEN

The plasticity of human adipose tissue-derived stem cells (hASCs) is promising, but differentiation in vitro toward endothelial cells is poorly understood. Flow cytometry demonstrated that hASCs isolated from excised fat tissue were positive for CD29, CD44, CD70, CD90, CD105, and CD166 and negative for the endothelial marker CD31, and the hematopoietic cell markers CD34 and CD133. hASCs differentiated into adipocytes after cultivation in adipogenic medium. Exposure of hASCs for 10 days under hypoxia (3% oxygen) in combination with leptin increased the percentage of CD31(+) endothelial cells as well as CD31, VE-Cadherin, Flk-1, Tie2, von Willebrand factor, and endothelial cell nitric oxide synthase mRNA expression. This was enhanced on co-incubation of vascular endothelial growth factor (VEGF) and leptin, whereas VEGF alone was not sufficient. Moreover, hASCs cultured on a matrigel surface under hypoxia/VEGF/leptin, showed a stable branching network. Hypoxic conditions significantly decreased apoptosis as evaluated by cleaved caspase-3, and increased prolyl hydroxylase domain 3 mRNA expression. Hypoxia increased expression of VEGF as well as leptin transcripts, which were significantly inhibited on co-incubation with either VEGF or leptin or a combination of both. Furthermore, leptin treatment of hypoxic cells increased the expression of the long/signaling form of the leptin receptor (ObRL), which was augmented on co-incubation with VEGF. The observed endothelial differentiation was dependent on the Akt pathway, as co-administration with Akt inhibitor abolished the observed effects. In conclusion, our data demonstrate that hASCs can be efficiently differentiated to endothelial cells by mimicking the hypoxic and pro-angiogenic microenvironment of adipose tissue.


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular , Endotelio Vascular/citología , Leptina/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Grasa Abdominal/citología , Anciano , Apoptosis , Biomarcadores/metabolismo , Hipoxia de la Célula , Células Cultivadas , Humanos , Persona de Mediana Edad , Neovascularización Fisiológica , Transcriptoma
8.
Stem Cells Dev ; 22(14): 1977-90, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23441986

RESUMEN

Embryonic stem (ES) cells are a powerful model for the development of cells responsible for the cellular immune response. Therefore, we analyzed the defense and phagocytic capacity of embryoid bodies (EBs) derived from ES cells using in the vitro inflammatory conditions caused by Escherichia coli. Further, we used this phagocytic activity to purify activated immune cells. Our data show that spontaneously differentiated 18-day-old EBs of the cell line CGR8 contained immune cells, which were positive for CD45, CD68, CD11b, F4/80, and CD19. Exposure of these EBs to E. coli with defined infection doses of bacterial colony-forming units (CFUs) led to a significant time-dependent reduction of CFUs, indicating the immune responses exerted by EBs. This was paralleled by an upregulation of inflammatory cytokines, that is, IL-1ß and TNF-α. Western blot analysis of infected EBs indicated an upregulation of CD14 and cytochrome b-245 heavy chain (NOX2). Silencing of NOX2 significantly reduced the antibacterial capacity of EBs, which was partially explained by reduction of F4/80-positive cells. To identify, isolate, and further cultivate phagocytic active cells from differentiated EBs, a cocultivation assay of differentiated ES cells with green fluorescent protein (GFP)-labeled E. coli was established. Colocalization of GFP-labeled E. coli with cells positive for CD45, CD68, and F4/80 revealed time-dependent phagocytotic uptake, which was underlined by colocalization with the LysoTracker-Red(®) dye as well as preincubation with cytochalasin D. In conclusion, a primitive immune response with efficient phagocytosis was responsible for the antibacterial capacity of differentiated EBs.


Asunto(s)
Cuerpos Embrioides/inmunología , Escherichia coli/crecimiento & desarrollo , Fagocitosis/inmunología , Aminas , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Diferenciación Celular , Línea Celular , Técnicas de Cocultivo , Recuento de Colonia Microbiana , Citocalasina D , Cuerpos Embrioides/citología , Escherichia coli/inmunología , Citometría de Flujo , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología
9.
Stem Cells Dev ; 20(12): 2163-75, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21470048

RESUMEN

Nitric oxide (NO) is a key regulator of cardiomyogenesis of embryonic stem (ES) cells. However, signaling pathways involving the energy sensor AMP-activated protein kinase (AMPK) and/or mammalian target of rapamycin (mTOR) resulting in NO generation and stimulation of cardiomyogenesis are currently not known. Herein, the role of AMPK- versus mTOR-regulated signaling pathways and the impact of NO for cardiomyogenesis of mouse ES cells were investigated. Activation of AMPK by 5-amino-4-imidazolecarboxamide riboside (AICAr) or metformin as well as inactivation of AMPK by compound C (Comp C), siRNA ablation of AMPKα2, or exogenous ATP stimulated cardiomyogenesis of ES cells. Inhibition of AMPK by Comp C resulted in phosphorylation of mTOR and generation of NO. NO generation was likewise achieved when AMPK was either activated by AICAr or mTOR was inhibited by rapamycin, suggesting that NO generation occurred by two mutually active parallel signaling pathways, one being AMPK dependent and mTOR independent (AICAr pathway) and the other being AMPK independent and mTOR dependent (Comp C pathway). Consequently, cardiomyogenesis as well as NO generation was completely abrogated when ES cells were cultivated in the presence of rapamycin and Comp C, which inhibit both signaling pathways. The impact of NO for cardiomyogenesis of ES cells was corroborated in experiments showing that the effects of Comp C on cardiomyogenesis of ES cells were abolished by the NO synthase inhibitors NG-monomethyl-l-arginine and N (G)-nitro-l-arginine methyl ester. In summary, our data demonstrate that NO generation downstream of AMPK and mTOR is activated by distinct, interacting signaling pathways that initiate cardiomyogenesis of ES cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Miocitos Cardíacos/citología , Óxido Nítrico/farmacología , Organogénesis , Serina-Treonina Quinasas TOR/metabolismo , Adenosina Trifosfato/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Modelos Biológicos , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Organogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología
10.
Cardiovasc Res ; 86(1): 37-44, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19965960

RESUMEN

AIMS: It is well established that leukotrienes (LTs), products of the 5-lipoxygenase (5-LO) pathway, participate in inflammatory tissue reactions and immune responses. In the present study, the impact of the 5-LO pathway on vasculogenesis of mouse embryonic stem (ES) cells was investigated. METHODS AND RESULTS: Immunohistochemistry studies demonstrated that 5-LO(+) cells first appeared at day 6 of embryoid body (EB) formation from ES cells. 5-LO(+)/CD68(+) as well as 5-LO(+)/CD45(+) cells were prominent at day 10 of EB differentiation. Real-time PCR and western blot analysis revealed all constituents of the 5-LO pathway. High performance liquid chromatography analyses indicated the synthesis of LTB(4) and LTD(4) in conformity with induction of the 5-LO pathway. Furthermore, Flk-1(+)/CD105(+) cells displayed calcium- (Ca(2+)) transients in response to LTB(4), whereas CD11b(+) cells responded to LTD(4). Treatment of EBs with LTB(4) and LTD(4) resulted in phosphorylation of the mitogen-activated protein kinase ERK1/2. Pharmacological inhibition of the 5-LO pathway and stable shRNA targeting of 5-LO-activating protein decreased capillary cell areas positive for PECAM-1. CONCLUSION: Our data demonstrate that the 5-LO pathway emerges early during ES cell differentiation into cells of the myeloid lineage and that LTs play an until now unrecognized role in vascular development of ES cells.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Proteínas Portadoras/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Activadoras de la 5-Lipooxigenasa , Animales , Araquidonato 5-Lipooxigenasa/genética , Proteínas Portadoras/genética , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Epóxido Hidrolasas/genética , Epóxido Hidrolasas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Leucotrieno B4/farmacología , Leucotrieno D4/farmacología , Leucotrienos/metabolismo , Linfocitos/citología , Linfocitos/fisiología , Macrófagos/citología , Macrófagos/fisiología , Proteínas de la Membrana/genética , Ratones , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/metabolismo , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Stem Cells Dev ; 19(5): 731-43, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19788349

RESUMEN

Electromagnetic fields (EMFs) are used to treat bone diseases. Herein, the effects of static EMFs on chondroosteogenesis and vasculogenesis of embryonic stem (ES) cells and bone mineralization of mouse fetuses were investigated. Treatment of differentiating ES cells with static EMFs (0.4-2 mT) stimulated vasculogenesis and chondro-osteogenesis and increased reactive oxygen species (ROS), which was abolished by the free radical scavengers trolox, 1,10-phenanthroline (phen), and the NAD(P)H oxidase inhibitor diphenylen iodonium (DPI). In contrast, EMFs of 10 mT field strength exerted inhibitory effects on vasculogenesis and chondro-osteogenesis despite robust ROS generation. EMFs of 1 mT and 10 mT increased and decreased vascular endothelial growth factor (VEGF) expression, respectively, which was abolished by DPI and radical scavengers. EMFs activated extracellular-regulated kinase 1/2 (ERK1/2), p38, and c-jun N-terminal kinase (JNK), which was sensitive to DPI treatment. The increase in VEGF by EMFs was inhibited by the ERK1/2 inhibitor U0126 but not by SB203580 and SP600125, which are p38 and JNK inhibitors, respectively, suggesting VEGF regulation by ERK1/2. Chondroosteogenesis and vasculogenesis of ES cells was blunted by trolox, DPI, and the VEGF receptor-2 (flk-1) antagonist SU5614. In mouse fetuses 1 mT EMFs increased and 10 mT EMFs decreased bone mineralization, which was abolished in the presence of trolox. Hence, EMFs induced chondro-osteogenesis and vasculogenesis in ES cells and bone mineralization of mouse fetuses by a ROS-dependent up-regulation of VEGF expression.


Asunto(s)
Campos Electromagnéticos , Células Madre Embrionarias/fisiología , Neovascularización Fisiológica , Osteogénesis , Especies Reactivas de Oxígeno/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Calcificación Fisiológica , Células Madre Embrionarias/metabolismo , Feto , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda