Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 13(7)2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916304

RESUMEN

The intracellular calcium concentration ([Ca2+]i) modulation plays a key role in the regulation of cellular growth and survival in normal cells and failure of [Ca2+]i homeostasis is involved in tumor initiation and progression. Here we showed that inhibition of Furin by its naturally occurring inhibitor the prodomain ppFurin in the MDA-MB-231 breast cancer cells resulted in enhanced store-operated calcium entry (SOCE) and reduced the cell malignant phenotype. Expression of ppFurin in a stable manner in MDA-MB-231 and the melanoma MDA-MB-435 cell lines inhibits Furin activity as assessed by in vitro digestion assays. Accordingly, cell transfection experiments revealed that the ppFurin-expressing cells are unable to adequately process the proprotein convertase (PC) substrates vascular endothelial growth factor C (proVEGF-C) and insulin-like growth factor-1 receptor (proIGF-1R). Compared to MDA-MB-435 cells, expression of ppFurin in MDA-MB-231 and BT20 cells significantly enhanced SOCE and induced constitutive Ca2+ entry. The enhanced SOCE is impaired by inhibition of Orai channels while the constitutive Ca2+ entry is attenuated by silencing or inhibition of TRPC6 or inhibition of Orai channels. Analysis of TRPC6 activation revealed its upregulated tyrosine phosphorylation in ppFurin-expressing MDA-MB-231 cells. In addition, while ppFurin had no effect on MDA-MB-435 cell viability, in MDA-MB-231 cells ppFurin expression reduced their viability and ability to migrate and enhanced their sensitization to the apoptosis inducer hydrogen peroxide and similar results were observed in BT20 cells. These findings suggest that Furin inhibition by ppFurin may be a useful strategy to interfere with Ca2+ mobilization, leading to breast cancer cells' malignant phenotype repression and reduction of their resistance to treatments.

2.
Biochim Biophys Acta Mol Cell Res ; 1867(11): 118808, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32781095

RESUMEN

Preclinical lung cancer models are essential for a basic understanding of lung cancer biology and its translation into efficient treatment options for affected patients. Lung cancer cell lines and xenografts derived directly from human lung tumors have proven highly valuable in fundamental oncology research and anticancer drug discovery. Both models inherently comprise advantages and caveats that have to be accounted for. Recently, we have enabled reliable in vitro culture techniques from lung cancer biopsies as Patients Lung Derived Tumoroids (PLDTs). This breakthrough provides the possibility of high-throughput drug screening covering the spectrum of lung cancer phenotypes seen clinically. We have adapted and optimized our in vitro three-dimensional model as a preclinical lung cancer model to recapitulate the tumor microenvironment (TME) using matrix reconstitution. Hence, we developed directly PLDTs to screen for chemotherapeutics and radiation treatment. This original model will enable precision medicine to become a reality, allowing a given patient sample to be screened for effective ex vivo therapeutics, aiming at tailoring of treatments specific to that individual. Hence, this tool can enhance clinical outcomes and avoid morbidity due to ineffective therapies.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/patología , Cultivo Primario de Células , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Pulmonares/patología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
3.
JCI Insight ; 5(14)2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32516140

RESUMEN

Apelin is a well-established mediator of survival and mitogenic signaling through the apelin receptor (Aplnr) and has been implicated in various cancers; however, little is known regarding Elabela (ELA/APELA) signaling, also mediated by Aplnr, and its role and the role of the conversion of its precursor proELA into mature ELA in cancer are unknown. Here, we identified a function of mTORC1 signaling as an essential mediator of ELA that repressed kidney tumor cell growth, migration, and survival. Moreover, sunitinib and ELA showed a synergistic effect in repressing tumor growth and angiogenesis in mice. The use of site-directed mutagenesis and pharmacological experiments provided evidence that the alteration of the cleavage site of proELA by furin induced improved ELA antitumorigenic activity. Finally, a cohort of tumors and public data sets revealed that ELA was only repressed in the main human kidney cancer subtypes, namely clear cell, papillary, and chromophobe renal cell carcinoma. Aplnr was expressed by various kidney cells, whereas ELA was generally expressed by epithelial cells. Collectively, these results showed the tumor-suppressive role of mTORC1 signaling mediated by ELA and established the potential use of ELA or derivatives in kidney cancer treatment.


Asunto(s)
Receptores de Apelina/genética , Apelina/genética , Carcinoma de Células Renales/genética , Hormonas Peptídicas/genética , Animales , Apelina/metabolismo , Calcio/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Furina/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Riñón/efectos de los fármacos , Riñón/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Transducción de Señal/efectos de los fármacos , Sunitinib/farmacología , Proteínas Supresoras de Tumor/genética
4.
Cancer Res ; 79(19): 5008-5021, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31358531

RESUMEN

Proprotein convertases (PC) activate precursor proteins that play crucial roles in various cancers. In this study, we investigated whether PC enzyme activity is required for expression of the checkpoint protein programmed cell death protein 1 (PD-1) on cytotoxic T lymphocytes (CTL) in colon cancer. Although altered expression of the PC secretory pathway was observed in human colon cancers, only furin showed highly diffuse expression throughout the tumors. Inhibition of PCs in T cells using the general protein-based inhibitor α1-PDX or the pharmacologic inhibitor Decanoyl-Arg-Val-Lys-Arg-chloromethylketone repressed PD-1 and exhausted CTLs via induction of T-cell proliferation and apoptosis inhibition, which improved CTL efficacy against microsatellite instable and microsatellite stable colon cancer cells. In vivo, inhibition of PCs enhanced CTL infiltration in colorectal tumors and increased tumor clearance in syngeneic mice compared with immunodeficient mice. Inhibition of PCs repressed PD-1 expression by blocking proteolytic maturation of the Notch precursor, inhibiting calcium/NFAT and NF-κB signaling, and enhancing ERK activation. These findings define a key role for PCs in regulating PD-1 expression and suggest targeting PCs as an adjunct approach to colorectal tumor immunotherapy. SIGNIFICANCE: Protein convertase enzymatic activity is required for PD-1 expression on T cells, and inhibition of protein convertase improves T-cell targeting of microsatellite instable and stable colorectal cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/19/5008/F1.large.jpg.


Asunto(s)
Neoplasias Colorrectales/inmunología , Receptor de Muerte Celular Programada 1/biosíntesis , Proproteína Convertasas/metabolismo , Linfocitos T Citotóxicos/metabolismo , Microambiente Tumoral/inmunología , Animales , Neoplasias Colorrectales/metabolismo , Xenoinjertos , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Linfocitos T Citotóxicos/inmunología
5.
Cancer Res ; 76(22): 6507-6519, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27634764

RESUMEN

The CXCL4 paralog CXCL4L1 is a less studied chemokine that has been suggested to exert an antiangiogenic function. However, CXCL4L1 is also expressed in patient tumors, tumor cell lines, and murine xenografts, prompting a more detailed analysis of its role in cancer pathogenesis. We used genetic and antibody-based approaches to attenuate CXCL4L1 in models of pancreatic ductal adenocarcinoma (PDAC). Mechanisms of expression were assessed in cell coculture experiments, murine, and avian xenotransplants, including through an evaluation of CpG methylation and mutation of critical CpG residues. CXCL4L1 gene expression was increased greatly in primary and metastatic PDAC. We found that myofibroblasts triggered cues in the tumor microenvironment, which led to induction of CXCL4L1 in tumor cells. CXCL4L1 expression was also controlled by epigenetic modifications at critical CpG islands, which were mapped. CXCL4L1 inhibited angiogenesis but also affected tumor development more directly, depending on the tumor cell type. In vivo administration of an mAb against CXCL4L1 demonstrated a blockade in the growth of tumors positive for CXCR3, a critical receptor for CXCL4 ligands. Our findings define a protumorigenic role in PDAC development for endogenous CXCL4L1, which is independent of its antiangiogenic function. Cancer Res; 76(22); 6507-19. ©2016 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Neoplasias Pancreáticas/genética , Receptores CXCR3/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Quimiocinas , Humanos , Ratones , Neovascularización Patológica , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Factor Plaquetario 4 , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood ; 127(7): 908-20, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26634301

RESUMEN

Apelin peptide and its receptor APJ are directly implicated in various physiological processes ranging from cardiovascular homeostasis to immune signaling. Here, we show that apelin is a key player in hemostasis with an ability to inhibit thrombin- and collagen-mediated platelet activation. Mice lacking apelin displayed a shorter bleeding time and a prothrombotic profile. Their platelets exhibited increased adhesion and a reduced occlusion time in venules, and displayed a higher aggregation rate after their activation by thrombin compared with wild-type platelets. Consequently, human and mouse platelets express apelin and its receptor APJ. Apelin directly interferes with thrombin-mediated signaling pathways and platelet activation, secretion, and aggregation, but not with ADP and thromboxane A2-mediated pathways. IV apelin administration induced excessive bleeding and prevented thrombosis in mice. Taken together, these findings suggest that apelin and/or APJ agonists could potentially be useful adducts in antiplatelet therapies and may provide a promising perspective for patients who continue to display adverse thrombotic events with current antiplatelet therapies.


Asunto(s)
Adipoquinas/metabolismo , Plaquetas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Adhesividad Plaquetaria , Transducción de Señal , Adipoquinas/genética , Adipoquinas/farmacología , Animales , Apelina , Receptores de Apelina , Hemorragia/inducido químicamente , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Trombina/genética , Trombina/metabolismo , Trombosis/genética , Trombosis/metabolismo , Trombosis/prevención & control , Tromboxano A2/genética , Tromboxano A2/metabolismo
7.
Oncotarget ; 6(28): 24922-34, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26325176

RESUMEN

IRE1α is an endoplasmic reticulum (ER)-resident transmembrane signaling protein and a cellular stress sensor. The protein harbors a cytosolic dual kinase/endoribonuclease activity required for adaptive responses to micro-environmental changes. In an orthotopic xenograft model of human glioma, invalidation of IRE1α RNase or/and kinase activities generated tumors with remarkably distinct phenotypes. Contrasting with the extensive angiogenesis observed in tumors derived from control cells, the double kinase/RNase invalidation reprogrammed mesenchymal differentiation of cancer cells and produced avascular and infiltrative glioblastomas with blood vessel co-option. In comparison, selective invalidation of IRE1α RNase did not compromise tumor angiogenesis but still elicited invasive features and vessel co-option. In vitro, IRE1α RNase deficient cells were also endowed with a higher ability to migrate. Constitutive activation of both enzymes led to wild-type-like lesions. The presence of IRE1α, but not its RNase activity, is therefore required for glioblastoma neovascularization, whereas invasion results only from RNase inhibition. In this model, two key mechanisms of tumor progression and cancer cell survival are functionally linked to IRE1α.


Asunto(s)
Neoplasias Encefálicas/enzimología , Endorribonucleasas/metabolismo , Glioblastoma/enzimología , Neovascularización Patológica/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Doxiciclina/farmacología , Endorribonucleasas/genética , Glioblastoma/irrigación sanguínea , Glioblastoma/tratamiento farmacológico , Humanos , Immunoblotting , Estimación de Kaplan-Meier , Ratones , Microscopía Confocal , Mutación , Invasividad Neoplásica , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Proteínas Serina-Treonina Quinasas/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
PLoS Genet ; 11(7): e1005325, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26132659

RESUMEN

Gliomas are a highly heterogeneous group of brain tumours that are refractory to treatment, highly invasive and pro-angiogenic. Glioblastoma patients have an average survival time of less than 15 months. Understanding the molecular basis of different grades of glioma, from well differentiated, low-grade tumours to high-grade tumours, is a key step in defining new therapeutic targets. Here we use a data-driven approach to learn the structure of gene regulatory networks from observational data and use the resulting models to formulate hypothesis on the molecular determinants of glioma stage. Remarkably, integration of available knowledge with functional genomics datasets representing clinical and pre-clinical studies reveals important properties within the regulatory circuits controlling low and high-grade glioma. Our analyses first show that low and high-grade gliomas are characterised by a switch in activity of two subsets of Rho GTPases. The first one is involved in maintaining normal glial cell function, while the second is linked to the establishment of multiple hallmarks of cancer. Next, the development and application of a novel data integration methodology reveals novel functions of RND3 in controlling glioma cell migration, invasion, proliferation, angiogenesis and clinical outcome.


Asunto(s)
Neoplasias Encefálicas/genética , Redes Reguladoras de Genes/genética , Glioma/genética , Invasividad Neoplásica/genética , Proteínas de Unión al GTP rho/genética , Apoptosis/genética , Neoplasias Encefálicas/patología , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica/genética , Glioma/patología , Células HEK293 , Humanos , Interferencia de ARN , ARN Interferente Pequeño
9.
Development ; 140(16): 3311-22, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863483

RESUMEN

Pluripotent mouse embryonic stem cells (mESCs), maintained in the presence of the leukemia inhibitory factor (LIF) cytokine, provide a powerful model with which to study pluripotency and differentiation programs. Extensive microarray studies on cultured cells have led to the identification of three LIF signatures. Here we focus on muscle ras oncogene homolog (MRAS), which is a small GTPase of the Ras family encoded within the Pluri gene cluster. To characterise the effects of Mras on cell pluripotency and differentiation, we used gain- and loss-of-function strategies in mESCs and in the Xenopus laevis embryo, in which Mras gene structure and protein sequence are conserved. We show that persistent knockdown of Mras in mESCs reduces expression of specific master genes and that MRAS plays a crucial role in the downregulation of OCT4 and NANOG protein levels upon differentiation. In Xenopus, we demonstrate the potential of Mras to modulate cell fate at early steps of development and during neurogenesis. Overexpression of Mras allows gastrula cells to retain responsiveness to fibroblast growth factor (FGF) and activin. Collectively, these results highlight novel conserved and pleiotropic effects of MRAS in stem cells and early steps of development.


Asunto(s)
Células Madre Embrionarias/enzimología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión al GTP Monoméricas/metabolismo , Xenopus laevis/embriología , Activinas/farmacología , Secuencia de Aminoácidos , Animales , Biomarcadores/metabolismo , Encéfalo/embriología , Encéfalo/enzimología , Secuencia Conservada , Inducción Embrionaria , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Gástrula/citología , Gástrula/efectos de los fármacos , Gástrula/enzimología , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factor Inhibidor de Leucemia/farmacología , Ratones , Datos de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/genética , Proteína Homeótica Nanog , Neurogénesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ovario/enzimología , Xenopus laevis/genética , Xenopus laevis/metabolismo , Proteínas ras
10.
Mol Cell Proteomics ; 12(8): 2293-312, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23674615

RESUMEN

In order to map the extracellular or membrane proteome associated with the vasculature and the stroma in an embryonic organism in vivo, we developed a biotinylation technique for chicken embryo and combined it with mass spectrometry and bioinformatic analysis. We also applied this procedure to implanted tumors growing on the chorioallantoic membrane or after the induction of granulation tissue. Membrane and extracellular matrix proteins were the most abundant components identified. Relative quantitative analysis revealed differential protein expression patterns in several tissues. Through a bioinformatic approach, we determined endothelial cell protein expression signatures, which allowed us to identify several proteins not yet reported to be associated with endothelial cells or the vasculature. This is the first study reported so far that applies in vivo biotinylation, in combination with robust label-free quantitative proteomics approaches and bioinformatic analysis, to an embryonic organism. It also provides the first description of the vascular and matrix proteome of the embryo that might constitute the starting point for further developments.


Asunto(s)
Proteínas Aviares/metabolismo , Embrión de Pollo/metabolismo , Membrana Corioalantoides/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Animales , Biotinilación , Línea Celular Tumoral , Membrana Corioalantoides/lesiones , Humanos , Intestino Delgado/embriología , Intestino Delgado/metabolismo , Riñón/embriología , Riñón/metabolismo , Hígado/embriología , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Proteoma
11.
Angiogenesis ; 16(1): 181-94, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23076651

RESUMEN

The clear cell subtype of renal carcinoma (CCRCC) is highly vascularized and despite a slow progression rate, it is potentially a highly aggressive tumor. Although a doubling of median progression-free survival in CCRCC patients treated by targeted therapies has been observed, the fact that tumors escape after anti-VEGF treatment suggests alternative pathways. The chick chorioallantoic membrane (CAM) is a well-established model, which allows in vivo studies of tumor angiogenesis and the testing of anti-angiogenic molecules. However, only a few data exist on CCRCC grafted onto CAM. We aimed to validate herein the CAM as a suitable model for studying the development of CCRCC and the interactions with the surrounding stroma. Our study uses both CCRCC cell lines and fresh tumor samples after surgical resection. We demonstrate that in both cases CCRCC can be grafted onto the CAM, to survive and to induce an angiogenic process. We further provide insights into the transcriptional regulation of the model by performing a differential analysis of tumor-derived and stroma-derived transcripts.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/genética , Microscopía Confocal , Microvasos/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Fenotipo , Regulación hacia Arriba/genética
12.
BMC Genomics ; 11: 495, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20840761

RESUMEN

BACKGROUND: Modern functional genomic approaches may help to better understand the molecular events involved in tissue morphogenesis and to identify molecular signatures and pathways. We have recently applied transcriptomic profiling to evidence molecular signatures in the development of the normal chicken chorioallantoic membrane (CAM) and in tumor engrafted on the CAM. We have now extended our studies by performing a transcriptome analysis in the "wound model" of the chicken CAM, which is another relevant model of tissue morphogenesis. RESULTS: To induce granulation tissue (GT) formation, we performed wounding of the chicken CAM and compared gene expression to normal CAM at the same stage of development. Matched control samples from the same individual were used. We observed a total of 282 genes up-regulated and 44 genes down-regulated assuming a false-discovery rate at 5% and a fold change > 2. Furthermore, bioinformatics analysis lead to the identification of several categories that are associated to organismal injury, tissue morphology, cellular movement, inflammatory disease, development and immune system. Endothelial cell data filtering leads to the identification of several new genes with an endothelial cell signature. CONCLUSIONS: The chick chorioallantoic wound model allows the identification of gene signatures and pathways involved in GT formation and neoangiogenesis. This may constitute a fertile ground for further studies.


Asunto(s)
Membrana Corioalantoides/metabolismo , Membrana Corioalantoides/patología , Perfilación de la Expresión Génica , Modelos Biológicos , Cicatrización de Heridas/genética , Animales , Embrión de Pollo , Análisis por Conglomerados , Biología Computacional , Regulación hacia Abajo/genética , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Tejido de Granulación/metabolismo , Tejido de Granulación/patología , Inmunohistoquímica , Hibridación in Situ , Regulación hacia Arriba/genética
13.
Cell Biol Int ; 33(6): 621-31, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19248835

RESUMEN

Stromal cells follow a vascular smooth muscle differentiation pathway. However, cell culture models performed from human bone marrow do not allow the obtention of a large proportion of highly differentiated smooth muscle cells (SMC) and their differentiation pathways remain unclear. We have characterized a new model of SMC differentiation from human bone marrow stromal cells by using different factors (bFGF, EGF, insulin and BMP-4). A relative homogeneous population of differentiated SMC was reproducibly obtained in short-term culture with high expression of SMC markers. Id gene expression was investigated and showed that (1) Id2 mRNA expression was upregulated during SMC differentiation without change of Id1 mRNA and (2) Id1 gene expression highly increased concomitantly with a decrease of SMC markers while Id2 mRNA was slightly modulated. Our data suggested that Id genes are potentially implicated in the differentiation pathway of human SMC from bone marrow.


Asunto(s)
Células de la Médula Ósea/citología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Miocitos del Músculo Liso/metabolismo , Adulto , Anciano , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Inmunofenotipificación , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Persona de Mediana Edad , Miocitos del Músculo Liso/citología , ARN Mensajero/metabolismo , Células del Estroma/citología
14.
Int J Dev Biol ; 51(8): 745-52, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17939122

RESUMEN

Transcription enhancer factors 1 (TEF-1 or TEAD) make a highly conserved family of eukaryotic DNA binding proteins that activate not only viral regulatory elements but muscle specific genes and are involved in several developmental processes. In this study, we report the identification and the expression pattern of NTEF-1 (TEAD1) and DTEF-1 (TEAD3), two members of this family in Xenopus laevis. Both X. laevis NTEF-1 (XNTEF-1 or XTEAD1) and DTEF-1 (XDTEF-1 or XTEAD3) possess a 72 amino acid TEA domain characteristic of TEF-1 proteins. XNTEF-1 is a 426 amino acid protein that has 96% identity with the avian or the mammalian NTEF-1 proteins while XDTEF-1 is a 433 amino acid protein with 77 to 80% identity with the avian and mammalian DTEF-1 sequences respectively. Temporal expression analysis by RT-PCR indicated that the two genes are expressed maternally and throughout embryonic development. In the adult, the two genes are broadly expressed although they showed differences of expression between tissues. Spatial expression analysis by whole mount in situ hybridization showed that the XNTEF-1 and XDTEF-1 mRNAS were predominantly detected in eye, embryonic brain, somites and heart. In animal cap assay, the two genes are activated by bFGF but are differently regulated by BMP4, and the muscle regulatory factor Mef2d.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/metabolismo , Xenopus laevis/metabolismo , Secuencia de Aminoácidos , Animales , ADN Complementario/metabolismo , Desarrollo Embrionario , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Proteína MioD/metabolismo , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Transcripción de Dominio TEA , Factores de Tiempo
15.
Exp Cell Res ; 312(18): 3539-45, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16938290

RESUMEN

Endocytosis is inhibited by overexpression of either dynamin-1 or dynamin-2 mutants because both isoforms form heterotetramers with endogenous dynamin-2 and interfere with its function. By contrast, other phenotypes, which are specifically triggered by overexpression of dynamin-2, but not dynamin-1 are likely to reflect endocytosis-independent, dynamin-2-specific functions and/or interactions. Using Dyn2/Dyn1 chimeras, we explored the structural requirements for a readily quantifiable, isoform-specific function of dynamin-2, the activation of caspase-3 to trigger apoptosis. Strikingly, swapping the highly homologous GTPase domain of dynamin-2 into dynamin-1 was sufficient to confer caspase-3 activation. Moreover, assembly-defective mutations in GED, dynamin's GAP/assembly domain, that inhibit endocytosis enhance caspase-3 activation. Thus, this dynamin-2-specific function is mechanistically distinct from and independent of its role in endocytosis. These findings have important implications for interpreting dynamin-2 dependent phenotypes in overexpression studies.


Asunto(s)
Dinamina II/química , Dinamina II/metabolismo , Endocitosis/fisiología , Conformación Proteica , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Dinamina I/química , Dinamina I/genética , Dinamina I/metabolismo , Dinamina II/genética , Activación Enzimática , Células HeLa , Humanos , Isoformas de Proteínas/química , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/genética , Transferrina/metabolismo
16.
J Biol Chem ; 280(27): 25604-10, 2005 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-15879597

RESUMEN

Fibroblast growth factor 2 (FGF-2) has been detected in the nuclei of many tissues and cell lines. Here we demonstrate that FGF-2 added exogenously to NIH3T3 cells enters the nucleus and interacts with the nuclear active 90-kDa ribosomal S6 kinase 2 (RSK2) in a cell cycle-dependent manner. By using purified proteins, FGF-2 is shown to directly interact through two separate domains with two RSK2 domains on both sides of the hydrophobic motif, namely the NH2-terminal kinase domain (residues 360-381) by amino acid Ser-117 and the COOH-terminal kinase domain (residues 388-400) by amino acids Leu-127 and Lys-128. Moreover, this interaction leads to maintenance of the sustained activation of RSK2 in G1 phase of the cell cycle. FGF-2 mutants (FGF-2 S117A, FGF-2 L127A, and FGF-2 K128A) that fail to interact in vitro with RSK2 fail to maintain a sustained RSK2 activity in vivo.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Fase S/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/genética , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Estructura Terciaria de Proteína , Fase de Descanso del Ciclo Celular/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/química , Fase S/fisiología
17.
Mol Biol Cell ; 16(4): 2058-67, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15703209

RESUMEN

Dynamin, a central player in clathrin-mediated endocytosis, interacts with several functionally diverse SH3 domain-containing proteins. However, the role of these interactions with regard to dynamin function is poorly defined. We have investigated a recently identified protein partner of dynamin, SNX9, sorting nexin 9. SNX9 binds directly to both dynamin-1 and dynamin-2. Moreover by stimulating dynamin assembly, SNX9 stimulates dynamin's basal GTPase activity and potentiates assembly-stimulated GTPase activity on liposomes. In fixed cells, we observe that SNX9 partially localizes to clathrin-coated pits. Using total internal reflection fluorescence microscopy in living cells, we detect a transient burst of EGFP-SNX9 recruitment to clathrin-coated pits that occurs during the late stages of vesicle formation and coincides spatially and temporally with a burst of dynamin-mRFP fluorescence. Transferrin internalization is inhibited in HeLa cells after siRNA-mediated knockdown of SNX9. Thus, our results establish that SNX9 is required for efficient clathrin-mediated endocytosis and suggest that it functions to regulate dynamin activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Clatrina/metabolismo , Dinamina II/metabolismo , Dinamina I/metabolismo , Endocitosis , Proteínas Portadoras/genética , Membrana Celular/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Dinamina I/química , Dinamina I/genética , Dinamina II/química , Dinamina II/genética , GTP Fosfohidrolasas/metabolismo , Células HeLa , Humanos , Liposomas/metabolismo , Unión Proteica , Receptores de Transferrina/metabolismo , Nexinas de Clasificación , Proteínas de Transporte Vesicular
18.
Cardiovasc Res ; 62(1): 154-66, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15023562

RESUMEN

OBJECTIVE: Fibroblast growth factor-2 (FGF-2), given during ischemia or during reperfusion of the ischemic heart is cardioprotective, but its mitogenic activity may limit possible clinical applications. We have tested the cardioprotective potential of a non-mitogenic FGF-2 mutant (S117A) that no longer activates casein kinase 2 (CK2) in both acute and long-term studies. METHODS AND RESULTS: To test effects during reperfusion, the ex vivo rat heart, subjected to 30 min of global ischemia and 60 min of reperfusion was used. S117A FGF-2 administered during reperfusion protected against myocardial contractile dysfunction, activated protein kinase C and decreased the release of cytochrome C in the cytosol. To study effects on ischemic myocytes in the absence of reperfusion, myocardial infarction (MI) was induced in the rat model by irreversible left coronary ligation. S117A-, wild type (wt)-FGF-2 or saline, were administered by intramyocardial injection into the ischemic ventricular wall. One day later, infarct size (assessed by tetrazolium staining), and plasma cardiac troponin T levels (assessed by Western blotting) were significantly decreased in the S117A FGF-2-, compared to the saline-treated group. Systolic pressure, rates of contraction and relaxation and developed pressure, assessed in the Langendorff mode, were significantly improved in the S117-FGF-2 group. Improved ejection fraction and fractional shortening in the S117A-treated group were maintained up to, but not beyond, 7 days post-MI. In comparison, improvements were maintained in the wt-FGF-2-treated group at least up to 6 weeks post-MI. At 6 weeks post-MI, small vessel density (assessed by immunofluorescence-based detection) in the scar bordering viable myocardium was similar between S117A-FGF-2- and saline-treated hearts, but significantly increased in the wt-FGF-2-treated group. This was accompanied by increased coronary flow in the wt-, but not S117A-FGF-2-treated hearts, compared to controls. CONCLUSION: The ability of FGF-2, administered during ischemia or during reperfusion, to protect the myocardium acutely from tissue loss and dysfunction is independent of its potential for CK2 activation and angiogenesis. Non-angiogenic S117A-FGF-2 may be considered in therapies aiming for acute prevention of reperfusion-associated pathologies, especially in cases where use of mitogens is counter-indicated.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Circulación Coronaria , Citocromos c/metabolismo , Ecocardiografía , Factor 2 de Crecimiento de Fibroblastos/genética , Masculino , Mutación , Isquemia Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA