Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
2.
Oncogene ; 31(18): 2270-82, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21927024

RESUMEN

Tumors are thought to be sustained by a reservoir of self-renewing cells, termed tumor-initiating cells or cancer stem cells. Osteosarcomas are high-grade sarcomas derived from osteoblast progenitor cells and are the most common pediatric bone malignancy. In this report we show that the stem cell transcription factor Sox2 is highly expressed in human and murine osteosarcoma (mOS) cell lines as well as in the tumor samples. Osteosarcoma cells have increased ability to grow in suspension as osteospheres, that are greatly enriched in expression of Sox2 and the stem cell marker, Sca-1. Depletion of Sox2 by short-hairpin RNAs in independent mOS-derived cells drastically reduces their transformed properties in vitro and their ability to form tumors. Sox2-depleted osteosarcoma cells can no longer form osteospheres and differentiate into mature osteoblasts. Concomitantly, they exhibit decreased Sca-1 expression and upregulation of the Wnt signaling pathway. Thus, despite other mutations, these cells maintain a requirement for Sox2 for tumorigenicity. Our data indicate that Sox2 is required for osteosarcoma cell self renewal, and that Sox2 antagonizes the pro-differentiation Wnt pathway that can in turn reduce Sox2 expression. These studies define Sox2 as a survival factor and a novel biomarker of self renewal in osteosarcomas, and support a tumor suppressive role for the Wnt pathway in tumors of mesenchymal origin. Our findings could provide the basis for novel therapeutic strategies based on inhibiting Sox2 or enhancing Wnt signaling for the treatment of osteosarcomas.


Asunto(s)
Neoplasias Óseas/genética , Proliferación Celular , Células Madre Neoplásicas , Osteosarcoma/genética , Factores de Transcripción SOXB1/genética , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Neoplasias Óseas/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Osteosarcoma/patología , Transducción de Señal , Vía de Señalización Wnt
3.
Cell Death Differ ; 17(8): 1345-53, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20489730

RESUMEN

The development and maintenance of most tissues and organs require the presence of multipotent and unipotent stem cells that have the ability of self-renewal as well as of generating committed, further differentiated cell types. The transcription factor Sox2 is essential for embryonic development and maintains pluripotency and self-renewal in embryonic stem cells. It is expressed in immature osteoblasts/osteoprogenitors in vitro and in vivo and is induced by fibroblast growth factor signaling, which stimulates osteoblast proliferation and inhibits differentiation. Sox2 overexpression can by itself inhibit osteoblast differentiation. To elucidate its function in the osteoblastic lineage, we generated mice with an osteoblast-specific, Cre-mediated knockout of Sox2. These mice are small and osteopenic, and mosaic for Sox2 inactivation. However, culturing calvarial osteoblasts from the mutant mice for 2-3 passages failed to yield any Sox2-null cells. Inactivation of the Sox2 gene by Cre-mediated excision in cultured osteoblasts showed that Sox2-null cells could not survive repeated passage in culture, could not form colonies, and arrested their growth with a senescent phenotype. In addition, expression of Sox2-specific shRNAs in independent osteoblastic cell lines suppressed their proliferative ability. Osteoblasts capable of forming 'osteospheres' are greatly enriched in Sox2 expression. These data identify a novel function for Sox2 in the maintenance of self-renewal in the osteoblastic lineage.


Asunto(s)
Osteoblastos/citología , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción Activador 2/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Desarrollo Embrionario , Ratones , Ratones Noqueados , Interferencia de ARN , Factores de Transcripción SOXB1/genética , Transducción de Señal
4.
Clin Infect Dis ; 33(9): 1579-85, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11588701

RESUMEN

A retrospective analysis of data from a cohort of patients coinfected with human immunodeficiency virus (HIV) and hepatitis C virus (HCV) who were treated with highly active antiretroviral therapy (HAART) at 3 infectious diseases units in northern Italy was performed. While the patients were receiving HAART, CD4(+) cell counts significantly increased and HIV RNA serum levels decreased. However, no significant overall changes in alanine aminotransferase (ALT) levels and HCV RNA serum levels were observed. Fifteen (4.6%) of 323 patients died within 3 years of follow-up; death was related to cirrhosis in 5 patients (1.6%). No significant difference was observed between cirrhosis-related mortality and mortality related to other causes. Patients with ALT levels >4 times the normal values at initiation of HAART showed a significant decrease in ALT levels, whereas patients with normal ALT levels at initiation of HAART showed a significant increase over time, suggesting that HAART may have long-term beneficial or detrimental effects, depending on patient characteristics.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , VIH/fisiología , Hepacivirus/fisiología , Hepatitis C/tratamiento farmacológico , Alanina Transaminasa/sangre , Terapia Antirretroviral Altamente Activa , Recuento de Linfocito CD4 , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , VIH/genética , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Hepacivirus/genética , Hepatitis C/complicaciones , Hepatitis C/inmunología , Hepatitis C/virología , Humanos , Cirrosis Hepática , Masculino , ARN Viral/sangre , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento , Carga Viral
5.
Mol Cell Biol ; 21(17): 5946-57, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11486033

RESUMEN

Fibroblast growth factors (FGFs) comprise a large family of multifunctional, heparin-binding polypeptides that show diverse patterns of interaction with a family of receptors (FGFR1 to -4) that are subject to alternative splicing. FGFR binding specificity is an essential mechanism in the regulation of FGF signaling and is achieved through primary sequence differences among FGFs and FGFRs and through usage of two alternative exons, IIIc and IIIb, for the second half of immunoglobulin-like domain 3 (D3) in FGFRs. While FGF4 binds and activates the IIIc splice forms of FGFR1 to -3 at comparable levels, it shows little activity towards the IIIb splice forms of FGFR1 to -3 as well as towards FGFR4. To begin to explore the structural determinants for this differential affinity, we determined the crystal structure of FGF4 at a 1.8-A resolution. FGF4 adopts a beta-trefoil fold similar to other FGFs. To identify potential receptor and heparin binding sites in FGF4, a ternary FGF4-FGFR1-heparin model was constructed by superimposing the FGF4 structure onto FGF2 in the FGF2-FGFR1-heparin structure. Mutation of several key residues in FGF4, observed to interact with FGFR1 or with heparin in the model, produced ligands with reduced receptor binding and concomitant low mitogenic potential. Based on the modeling and mutational data, we propose that FGF4, like FGF2, but unlike FGF1, engages the betaC'-betaE loop in D3 and thus can differentiate between the IIIc and IIIb splice isoforms of FGFRs for binding. Moreover, we show that FGF4 needs to interact with both the 2-O- and 6-O-sulfates in heparin to exert its optimal biological activity.


Asunto(s)
Factores de Crecimiento de Fibroblastos/química , Heparina/química , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/química , Receptores de Factores de Crecimiento de Fibroblastos/química , Células 3T3 , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Cricetinae , Factor 4 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos
6.
Development ; 128(11): 2119-29, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11493533

RESUMEN

Unregulated FGF receptor signaling results in bone malformations that affect both endochondral and intramembranous ossification, and is the basis for several genetic forms of human dwarfism. FGF signaling inhibits chondrocyte proliferation and we have previously shown that the transcription factor STAT1 mediates the growth inhibitory effect of FGF in vitro. We provide genetic evidence that STAT1 is a modulator of the negative regulation of bone growth by FGF in vivo. We crossed Stat1(-/-) mice with a transgenic mouse line overexpressing human FGF2 (TgFGF). TgFGF mice exhibit phenotypes characterized by chondrodysplasia and macrocephaly, which affect endochondral and intramembranous ossification. We found that the chondrodysplasic phenotype of these mice results both from reduced proliferation and increased apoptosis of growth plate chondrocytes. Loss of STAT1 function in TgFGF mice led to a significant correction of the chondrodysplasic phenotype, but did not affect the skull malformations. The reduced proliferation of TgFGF growth plate chondrocytes, as well as their excessive apoptosis, were restored to near-normal levels in the absence of STAT1 function. Unregulated FGF signaling in TgFGF mice also induced apoptosis in calvarial osteoblasts that was not, however, corrected by the absence of STAT1. Detailed analysis of Stat1(-/-) growth plates uncovered a transient phenotype, characterized by an expansion of the proliferative zone and by acceleration of longitudinal bone growth, that attenuated as the animals grew older. These results document an essential role for STAT1 in FGF-mediated regulation of cell growth that is specific to the epiphyseal growth plate.


Asunto(s)
Apoptosis , Desarrollo Óseo , Condrocitos/citología , Proteínas de Unión al ADN/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Transactivadores/fisiología , Animales , División Celular , Proteínas de Unión al ADN/genética , Enanismo/etiología , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteocondrodisplasias/etiología , Fenotipo , Factor de Transcripción STAT1 , Cráneo/citología , Transactivadores/genética
7.
Eur Cytokine Netw ; 12(2): 361-4, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11399527

RESUMEN

Interleukin (IL)-18, a newly discovered cytokine produced primarily by macrophages, has been shown to induce gamma interferon (IFN-gamma) production by natural killer cells, to induce the T helper type 1 response. To further elucidate the role of this cytokine in uncomplicated malaria caused by Plasmodium falciparum, serum levels of IL-18, and gamma interferon (IFN-gamma), determined by an immunoenzymatic assay, were analyzed in 40 adult patients, and in 15 healthy control subjects. A significant increase in serum levels of IL-18 was observed in patients with uncomplicated P. falciparum malaria on admission, whereas serum levels of IFN-gamma tended to increase although not significantly. Serum levels of IL-18 decreased three days later, but still remained significantly high, whereas IFN-gamma levels returned to normal levels compared to the controls. No significant correlation was found between parasitemia and serum levels of IL-18 and IFN-gamma. The increase of IL-18 levels during acute and recovery phases of uncomplicated P. falciparum malaria may reflect a proinflammatory role of IL-18 in these patients. An early and effective immune response regulated by proinflammatory Th1 cytokines, including tumor necrosis factor (TNF), interleukin (IL)-12, and possibly IFN-gamma may limit the progression from uncomplicated malaria to severe and life-threatening complications.


Asunto(s)
Interleucina-18/sangre , Malaria Falciparum/sangre , Adulto , Femenino , Humanos , Técnicas para Inmunoenzimas , Interferón gamma/sangre , Masculino
9.
J Cell Physiol ; 186(3): 315-28, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11169970

RESUMEN

The highly conserved homeodomains and HMG domains are components of a large number of proteins that play a role in the transcriptional regulation of gene expression during embryogenesis. Both the HMG domain and the homeodomain serve as interfaces for factor interactions with DNA, as well as with other proteins, and it is likely that the high degree of structural and sequence conservation within these domains reflects the conservation of basic aspects of these interactions. Classical HMG domain proteins have an ancient origin, being found in all eukaryotes, and are thought to have given rise to the metazoan-specific class of HMG domain proteins called the Sox proteins. Similarly, the metazoan-specific POU domain proteins are thought to have arisen from genes encoding ancestral homeodomain proteins. In this review, we summarize several examples of different HMG-homeodomain interactions that illustrate not only the ancient origin of each of these protein families, but also their relationship to each other, and discuss how coevolution of HMG and homeodomains may have lead to creation of the specialized Sox/POU protein complexes. Using the FGF-4 gene as an example, we also speculate on how coevolution of regulatory Sox/POU target DNA sequences may have occurred, and how the summation of these changes may have lead to the emergence of new developmental pathways.


Asunto(s)
Evolución Molecular , Regulación de la Expresión Génica/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Proteínas de Homeodominio/genética , Filogenia , Transcripción Genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Secuencia Conservada , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
11.
Dev Biol ; 225(2): 392-406, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10985858

RESUMEN

The Fgf4 gene encodes an important signaling molecule which is expressed in specific developmental stages, including the inner cell mass of the blastocyst, the myotomes, and the limb bud apical ectodermal ridge (AER). Using a transgenic approach, we previously identified overlapping but distinct enhancer elements in the Fgf4 3' untranslated region necessary and sufficient for myotome and AER expression. Here we have investigated the hypothesis that Fgf4 is a target of myogenic bHLH factors. We show by mutational analysis that a conserved E box located in the Fgf4 myotome enhancer is required for Fgf4-lacZ expression in the myotomes. A DNA probe containing the E box binds MYF5, MYOD, and bHLH-like activities from nuclear extracts of differentiating C2-7 myoblast cells, and both MYF5 and MYOD can activate gene expression of reporter plasmids containing the E-box element. Analyses of Myf5 and MyoD knockout mice harboring Fgf4-lacZ transgenes show that Myf5 is required for Fgf4 expression in the myotomes, while MyoD is not, but MyoD can sustain Fgf4 expression in the ventral myotomes in the absence of Myf5. Sonic hedgehog (Shh) signaling has been shown to have an essential inductive function in the expression of Myf5 and MyoD in the epaxial myotomes, but not in the hypaxial myotomes. We show here that expression of an Fgf4-lacZ transgene in Shh-/- embryos is suppressed not only in the epaxial but also in the hypaxial myotomes, while it is maintained in the AER. This suggests that Shh mediates Fgf4 activation in the myotomes through mechanisms independent of its role in the activation of myogenic factors. Thus, a cascade of events, involving Shh and bHLH factors, is responsible for activating Fgf4 expression in the myotomes in a spatial- and temporal-specific manner.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario y Fetal , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/embriología , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Transactivadores , Factores de Transcripción/metabolismo , Regiones no Traducidas 3'/genética , Células 3T3 , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Sitios de Unión , Blastocisto/fisiología , Cloranfenicol O-Acetiltransferasa/análisis , Cloranfenicol O-Acetiltransferasa/genética , Ectodermo/fisiología , Inducción Embrionaria , Elementos de Facilitación Genéticos , Factor 4 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/metabolismo , Células HeLa , Proteínas Hedgehog , Secuencias Hélice-Asa-Hélice , Humanos , Esbozos de los Miembros/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes de Fusión/genética , Transfección , beta-Galactosidasa/genética
12.
J Cell Biol ; 149(6): 1297-308, 2000 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-10851026

RESUMEN

Fibroblast growth factors (FGF) play a critical role in bone growth and development affecting both chondrogenesis and osteogenesis. During the process of intramembranous ossification, which leads to the formation of the flat bones of the skull, unregulated FGF signaling can produce premature suture closure or craniosynostosis and other craniofacial deformities. Indeed, many human craniosynostosis disorders have been linked to activating mutations in FGF receptors (FGFR) 1 and 2, but the precise effects of FGF on the proliferation, maturation and differentiation of the target osteoblastic cells are still unclear. In this report, we studied the effects of FGF treatment on primary murine calvarial osteoblast, and on OB1, a newly established osteoblastic cell line. We show that FGF signaling has a dual effect on osteoblast proliferation and differentiation. FGFs activate the endogenous FGFRs leading to the formation of a Grb2/FRS2/Shp2 complex and activation of MAP kinase. However, immature osteoblasts respond to FGF treatment with increased proliferation, whereas in differentiating cells FGF does not induce DNA synthesis but causes apoptosis. When either primary or OB1 osteoblasts are induced to differentiate, FGF signaling inhibits expression of alkaline phosphatase, and blocks mineralization. To study the effect of craniosynostosis-linked mutations in osteoblasts, we introduced FGFR2 carrying either the C342Y (Crouzon syndrome) or the S252W (Apert syndrome) mutation in OB1 cells. Both mutations inhibited differentiation, while dramatically inducing apoptosis. Furthermore, we could also show that overexpression of FGF2 in transgenic mice leads to increased apoptosis in their calvaria. These data provide the first biochemical analysis of FGF signaling in osteoblasts, and show that FGF can act as a cell death inducer with distinct effects in proliferating and differentiating osteoblasts.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Osteoblastos/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Acrocefalosindactilia/genética , Fosfatasa Alcalina/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Calcificación Fisiológica/genética , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Disostosis Craneofacial/genética , Fragmentación del ADN/genética , Factor 1 de Crecimiento de Fibroblastos , Histocitoquímica , Ratones , Ratones Transgénicos , Mutación Puntual , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transfección
13.
J Biol Chem ; 275(30): 23387-97, 2000 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-10801796

RESUMEN

Fibroblast growth factor (FGF)-4 gene expression in the inner cell mass of the blastocyst and in EC cells requires the combined activity of two transcriptional regulators, Sox2 and Oct-3, which bind to adjacent sites on the FGF-4 enhancer DNA and synergistically activate transcription. Sox2 and Oct-3 bind cooperatively to the enhancer DNA through their DNA-binding, high mobility group and POU domains, respectively. These two domains, however, are not sufficient to activate transcription. We have analyzed a number of Sox2 and Oct-3 deletion mutants to identify the domains within each protein that contribute to the activity of the Sox2 x Oct-3 complex. Within Oct-3, we have identified two activation domains, the N-terminal AD1 and the C-terminal AD2, that play a role in the activity of the Sox2 x Oct-3 complex. AD1 also displays transcriptional activation functions in the absence of Sox2 while AD2 function was only detected within the Sox2 x Oct-3 complex. In Sox2, we have identified three activation domains within its C terminus: R1, R2, and R3. R1 and R2 can potentiate weak activation by Sox2 in the absence of Oct-3 but their deletion has no effect on the Sox2 x Oct-3 complex. In contrast, R3 function is only observed when Sox2 is complexed with Oct-3. In addition, analysis of Oct-1/Oct-3 chimeras indicates that the Oct-3 homeodomain also plays a critical role in the formation of a functional Sox2 x Oct-3 complex. Our results are consistent with a model in which the synergistic action of Sox2 and Oct-3 results from two major processes. Cooperative binding of the factors to the enhancer DNA, mediated by their binding domains, stably tethers each factor to DNA and increases the activity of intrinsic activation domains within each protein. Protein-protein and protein-DNA interactions then may lead to reciprocal conformational changes that expose latent activation domains within each protein. These findings define a mechanism that may also be utilized by other Sox x POU protein complexes in gene activation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Elementos de Facilitación Genéticos , Factores de Crecimiento de Fibroblastos/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Secuencia de Bases , Sitios de Unión , Cartilla de ADN , Factor 4 de Crecimiento de Fibroblastos , Regulación de la Expresión Génica , Proteínas HMGB , Células HeLa , Humanos , Factor 3 de Transcripción de Unión a Octámeros , Factores de Transcripción SOXB1
14.
Mol Cell Biol ; 20(6): 2260-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10688672

RESUMEN

Fibroblast growth factor 1 (FGF1) and FGF2, the prototypic members of the FGF family of growth factors, have been implicated in a variety of physiological and pathological processes. Unlike most other FGFs, FGF1 and FGF2 are ubiquitously expressed and are not efficiently secreted. Gene knockouts in mice have previously demonstrated a role for FGF2 in brain development, blood pressure regulation, and wound healing. The relatively mild phenotypic defects associated with FGF2 deletion led to the hypothesis that the continued expression of other FGFs partially compensated for the absence of FGF2 in these mice. We now report our generation of mice lacking FGF1 and their use, in combination with our previously described FGF2 null mice, to produce mice lacking both FGF1 and FGF2. FGF1-FGF2 double-knockout mice are viable and fertile and do not display any gross phenotypic defects. In the double-knockout mice we observed defects that were similar in extent to those previously described for the FGF2 null mice. Differences in the organization of neurons of the frontal motor cortex and in the rates of wound healing were observed. We also observed in FGF2(-/-) mice and in FGF1-FGF2 double-knockout mice novel impairments in hematopoiesis that were similar in severity. Essentially no abnormalities were found in mice lacking only FGF1. Our results suggest that the relatively mild defects in FGF2 knockout animals are not a consequence of compensation by FGF1 and suggest highly restricted roles for both factors under normal developmental and physiological conditions.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/genética , Animales , Desarrollo Embrionario y Fetal/genética , Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados
15.
FASEB J ; 14(2): 399-406, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10657996

RESUMEN

The MET proto-oncogene, encoding the tyrosine kinase receptor for HGF, controls genetic programs leading to cell growth, invasiveness, and protection from apoptosis. Recently, MET mutations have been identified in hereditary and sporadic forms of papillary renal carcinoma (PRC). Introduction of different naturally occurring mutations into the MET cDNA results in the acquisition of distinct biochemical and biological properties of transfected cells. Some mutations result in a high increase in tyrosine kinase activity and confer transforming ability in focus forming assays. These mutants hyperactivate the Ras signaling pathway. Other mutations are devoid of transforming potential but are effective in inducing protection from apoptosis and sustaining anchorage-independent growth. These Met(PRC) receptors interact more efficiently with the intracellular transducer Pi3Kinase. The reported results show that MET(PRC) mutations can be responsible for malignant transformation through different mechanisms, either by increasing the growth ability of cells or by protecting cells from apoptosis and allowing accumulation of other genetic lesions.-Giordano, S., Maffe, A., Williams, T. A., Artigiani, S., Gual, P., Bardelli, A., Basilico, C., Michieli, P., Comoglio, P. M. Different point mutations in the met oncogene elicit distinct biological properties.


Asunto(s)
Transformación Celular Neoplásica/genética , Mutación Puntual , Proteínas Proto-Oncogénicas c-met/genética , Apoptosis/genética , Adhesión Celular/genética , Movimiento Celular/genética , Células Epiteliales , Transducción de Señal/genética , Proteínas ras/metabolismo
16.
Microbes Infect ; 2(15): 1831-3, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11165927

RESUMEN

Kaposi's sarcoma (KS) is an angioproliferative disease characterized by proliferation of neoplastic cells (spindle cells) mixed with endothelial and inflammatory cells. In this study we evaluated the role of the adhesive glycoprotein, fibronectin (FN) and its receptor alpha(5)beta(1) (FNR), and the proto-oncogene bcl-2, an anti-apoptotic protein. Significantly decreased serum levels of FN were noted in HIV-1-infected patients with KS, whereas serum levels of FNR were significantly increased in the same patients. Furthermore, increased FNR expression was observed on CD4 cells from KS patients. Serum levels of bcl-2 protein were significantly decreased in asymptomatic seropositive patients, whereas HIV-1-infected patients with KS showed increased serum levels of bcl-2. These results provide further information about interaction between integrins and the extracellular matrix and bcl-2 protein that can support cell survival either of neoplastic cells or endothelial and inflammatory cells.


Asunto(s)
Infecciones Oportunistas Relacionadas con el SIDA/sangre , Fibronectinas/sangre , VIH-1 , Proteínas Proto-Oncogénicas c-bcl-2/sangre , Receptores de Fibronectina/sangre , Sarcoma de Kaposi/sangre , Infecciones Oportunistas Relacionadas con el SIDA/virología , Adulto , Linfocitos T CD4-Positivos/metabolismo , Femenino , Humanos , Masculino , Proto-Oncogenes Mas , Sarcoma de Kaposi/virología
17.
Oncogene ; 18(37): 5221-31, 1999 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-10498872

RESUMEN

Mutations in the genes encoding for Met, Ret and Kit receptor tyrosine kinases invariably result in increased kinase activity and in the acquisition of transforming potential. However, the requirement of receptor ligands for the transformation process is still unclear. We have investigated the role of hepatocyte growth factor (HGF), the high-affinity ligand for Met, in mutant Met-mediated cell transformation. We provide evidence that the transforming potential displayed by mutant forms of Met found in human cancer is not only sensitive but entirely dependent on the presence of HGF, by showing that mutant Met transforms NIH3T3 fibroblasts, which produce endogenous HGF, but is not able to transform epithelial cells, unless exogenous HGF is supplied. Accordingly, mutant Met-induced transformation of NIH3T3 cells can be inhibited by HGF antagonists and increased by HGF stimulation. We also show that an engineered Met receptor which contains an oncogenic mutation but is impaired in its ability to bind HGF completely loses its transforming activity, which can be rescued by causing receptor dimerization using a monoclonal antibody. These results indicate that point mutations resulting in Met kinase activation are necessary but not sufficient to cause cell transformation, the latter being dependent on ligand-induced receptor dimerization. They also suggest that mutant Met-driven tumour growth depends on the availability and tissue distribution of active HGF, and provide proof-of-concept for the treatment of mutant-Met related pathologies by HGF-antagonizing drugs.


Asunto(s)
Transformación Celular Neoplásica/genética , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/fisiología , Células 3T3 , Sustitución de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/clasificación , Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Dimerización , Diseño de Fármacos , Células Epiteliales , Factor de Crecimiento de Hepatocito/inmunología , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Ligandos , Ratones , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-met/genética , Proto-Oncogenes , Proteínas Recombinantes de Fusión/fisiología , Transfección , Ensayo de Tumor de Célula Madre
18.
Genes Dev ; 13(11): 1361-6, 1999 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10364154

RESUMEN

Several genetic forms of human dwarfism have been linked to activating mutations in FGF receptor 3, indicating that FGF signaling has a critical role in chondrocyte maturation and skeletal development. However, the mechanisms through which FGFs affect chondrocyte proliferation and differentiation remain poorly understood. We show here that activation of FGF signaling inhibits chondrocyte proliferation both in a rat chondrosarcoma (RCS) cell line and in primary murine chondrocytes. FGF treatment of RCS cells induces phosphorylation of STAT-1, its translocation to the nucleus, and an increase in the expression of the cell-cycle inhibitor p21WAF1/CIP1. We have used primary chondrocytes from STAT-1 knock-out mice to provide genetic evidence that STAT-1 function is required for the FGF mediated growth inhibition. Furthermore, FGF treatment of metatarsal rudiments from wild-type and STAT-1(-/-) murine embryos produces a drastic impairment of chondrocyte proliferation and bone development in wild-type, but not in STAT-1(-/-) rudiments. We propose that STAT-1 mediated down regulation of chondrocyte proliferation by FGF signaling is an homeostatic mechanism which ensures harmonious bone development and morphogenesis.


Asunto(s)
Desarrollo Óseo , Condrocitos/citología , Proteínas de Unión al ADN/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Células 3T3 , Animales , Desarrollo Óseo/efectos de los fármacos , Huesos/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Proteínas de Unión al ADN/genética , Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Huesos Metatarsianos/efectos de los fármacos , Huesos Metatarsianos/crecimiento & desarrollo , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fosforilación , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transactivadores/genética , Células Tumorales Cultivadas
19.
Mol Endocrinol ; 13(2): 191-201, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9973250

RESUMEN

We identified Ark, the mouse homolog of the receptor tyrosine kinase Axl (Ufo, Tyro7), in a screen for novel factors involved in GnRH neuronal migration by using differential-display PCR on cell lines derived at two windows during GnRH neuronal development. Ark is expressed in Gn10 GnRH cells, developed from a tumor in the olfactory area when GnRH neurons are migrating, but not in GT1-7 cells, derived from a tumor in the forebrain when GnRH neurons are postmigratory. Since Ark (Ax1) signaling protects from programmed cell death in fibroblasts, we hypothesized that it may play an antiapoptotic role in GnRH neurons. Gn10 (Ark positive) GnRH cells were more resistant to serum withdrawal-induced apoptosis than GT1-7 (Ark negative) cells, and this effect was augmented with the addition of Gas6, the Ark (Ax1) ligand. Gas6/Ark stimulated the extracellular signal-regulated kinase, ERK, and the serine-threonine kinase, Akt, a downstream component of the phosphoinositide 3-kinase (PI3-K) pathway. To determine whether ERK or Akt activation is required for the antiapoptotic effects of Gas6/Ark in GnRH neurons, cells were serum starved in the absence or presence of Gas6, with or without inhibitors of ERK and PI3-K signaling cascades. Gas6 rescued Gn10 cells from apoptosis, and this effect was blocked by coincubation of the cells with the mitogen-activated protein/ERK kinase (MEK) inhibitor, PD98059, or wortmannin (but not rapamycin). These data support an important role for Gas6/Ark signaling via the ERK and PI3-K (via Akt) pathways in the protection of GnRH neurons from programmed cell death across neuronal migration.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Neuronas/fisiología , Proteínas Oncogénicas , Proteínas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Androstadienos/farmacología , Animales , Apoptosis/fisiología , Northern Blotting , Western Blotting , Bromodesoxiuridina/química , Comunicación Celular/fisiología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Flavonoides/farmacología , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunosupresores/farmacología , Ratones , Microscopía Fluorescente , Pruebas de Precipitina , Proteínas/genética , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/genética , Sirolimus/farmacología , Wortmanina , Tirosina Quinasa del Receptor Axl
20.
Dev Biol ; 204(1): 197-209, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9851853

RESUMEN

Embryonic development requires a complex program of events which are directed by a number of signaling molecules whose expression must be rigorously regulated. We previously showed that expression of Fgf4, which plays an important role in postimplantation development and growth and patterning of the limb, is regulated in EC cells by the synergistic interaction of Sox2 and Oct-3 with the Fgf4 EC cell-specific enhancer. To verify whether this mechanism was also operating in vivo, and to identify new elements controlling Fgf4 gene expression in distinct developmental stages, we have analyzed the expression of LacZ reporter plasmids containing different fragments of the Fgf4 gene in transgenic mouse embryos. Utilizing these transgenic constructs we have been able to recapitulate, for the most part, Fgf4 gene expression during embryonic development. We show here that most of the cis-acting regulatory elements determining Fgf4 embryonic expression are located in conserved regions within the 3' UTR of the gene. The EC cell-specific enhancer is required to drive gene expression in the ICM of the blastocyst, and its activity requires the Sox and Oct-proteins binding sites. We were also able to identify specific and distinct enhancer elements that govern postimplantation expression in the somitic myotomes and the limb bud AER. The myotome-specific elements contain binding sites for bHLH myogenic regulatory factors, which appear to be essential for myotome expression. Finally, we present evidence that the very restricted pattern of expression of Fgf4 transcripts in the AER results from the combined action of positive and negative regulatory elements located 3' of the Fgf4 coding sequences. Thus the Fgf4 gene relies on multiple and distinct regulatory elements to achieve stage- and tissue-specific embryonic expression.


Asunto(s)
Blastocisto , Extremidades/embriología , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Animales , Desarrollo Embrionario y Fetal/genética , Factor 4 de Crecimiento de Fibroblastos , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA