Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
BMC Genomics ; 16: 986, 2015 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-26589636

RESUMEN

BACKGROUND: The genetic program, as manifested as the cellular phenotype, is in large part dictated by the cell's protein composition. Since characterisation of the proteome remains technically laborious it is attractive to define the genetic expression profile using the transcriptome. However, the transcriptional landscape is complex and it is unclear as to what extent it reflects the ribosome associated mRNA population (the translatome). This is particularly pertinent for genes using multiple transcriptional start sites (TSS) generating mRNAs with heterogeneous 5' transcript leaders (5'TL). Furthermore, the relative abundance of the TSS gene variants is frequently cell-type specific. Indeed, promoter switches have been reported in pathologies such as cancer. The consequences of this 5'TL heterogeneity within the transcriptome for the translatome remain unresolved. This is not a moot point because the 5'TL plays a key role in regulating mRNA recruitment onto polysomes. RESULTS: In this article, we have characterised both the transcriptome and translatome of the MCF7 (tumoural) and MCF10A (non-tumoural) cell lines. We identified ~550 genes exhibiting differential translation efficiency (TE). In itself, this is maybe not surprising. However, by focusing on genes exhibiting TSS heterogeneity we observed distinct differential promoter usage patterns in both the transcriptome and translatome. Only a minor fraction of these genes belonged to those exhibiting differential TE. Nonetheless, reporter assays demonstrated that the TSS variants impacted on the translational readout both quantitatively (the overall amount of protein expressed) and qualitatively (the nature of the proteins expressed). CONCLUSIONS: The results point to considerable and distinct cell-specific 5'TL heterogeneity within both the transcriptome and translatome of the two cell lines analysed. This observation is in-line with the ribosome filter hypothesis which posits that the ribosomal machine can selectively filter information from within the transcriptome. As such it cautions against the simple extrapolation transcriptome → proteome. Furthermore, polysomal occupancy of specific gene 5'TL variants may also serve as novel disease biomarkers.


Asunto(s)
Polirribosomas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Sitio de Iniciación de la Transcripción , Animales , Línea Celular , Biología Computacional/métodos , Regulación de la Expresión Génica , Humanos , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , Regiones Promotoras Genéticas , Caperuzas de ARN , ARN Mensajero/química , Transcriptoma
2.
J Cell Physiol ; 229(11): 1765-75, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24664975

RESUMEN

Age-related bone loss is characterized by reduced osteoblastogenesis and excessive bone marrow adipogenesis. The mechanisms governing bone marrow mesenchymal stromal cell (BMSC) differentiation into adipocytes or osteoblasts during aging are unknown. We show here that overexpressing N-cadherin (Cadh2) in osteoblasts increased BMSC adipocyte differentiation and reduced osteoblast differentiation in young transgenic (Tg) mice whereas this phenotype was fully reversed with aging. The reversed phenotype with age was associated with enhanced Wnt5a and Wnt10b expression in osteoblasts and a concomitant increase in BMSC osteogenic differentiation. Consistent with this mechanism, conditioned media from young wild type osteoblasts inhibited adipogenesis and promoted osteoblast differentiation in BMSC from old Cadh2 Tg mice, and this response was abolished by Wnt5a and Wnt10b silencing. Transplantation of BMSC from old Cadh2 Tg mice into young Tg recipients increased Wnt5a and Wnt10b expression and rescued BMSC osteogenic differentiation. In senescent osteopenic mice, blocking the CADH2-Wnt interaction using an antagonist peptide increased Wnt5a and Wnt10b expression, bone formation, and bone mass. The data indicate that Cadh2/Wnt interaction in osteoblasts regulates BMSC lineage determination, bone formation, and bone mass and suggest a therapeutic target for promoting bone formation in the aging skeleton.


Asunto(s)
Envejecimiento/metabolismo , Células de la Médula Ósea/citología , Cadherinas/metabolismo , Linaje de la Célula , Células Madre Mesenquimatosas/citología , Proteínas Wnt/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Animales , Células de la Médula Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Tamaño de los Órganos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis , Unión Proteica , Transducción de Señal , Trasplante de Células Madre , Proteína Wnt-5a
3.
Gene ; 521(1): 55-61, 2013 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-23541807

RESUMEN

Osteosarcoma is the most common primary bone tumor in children and adults. Despite improved prognosis, resistance to chemotherapy remains responsible for failure of osteosarcoma treatment. The identification of the molecular signals that contribute to the aberrant osteosarcoma cell growth may provide clues to develop new therapeutic strategies for chemoresistant osteosarcoma. Here we show that the expression of ErbB3 is increased in human osteosarcoma cells in vitro. Tissue microarray analysis of tissue cores from osteosarcoma patients further showed that the ErbB3 protein expression is higher in bone tumors compared to normal bone tissue, and is further increased in patients with recurrent disease or soft tissue metastasis. In murine osteosarcoma cells, silencing ErbB3 using shRNA decreased cell replication, cell migration and invasion, indicating that ErbB3 contributes to tumor cell growth and invasiveness. Furthermore, ErbB3 silencing markedly reduced tumor growth in a murine allograft model in vivo. Immunohistochemal analysis showed that the reduced tumor growth induced by ErbB3 silencing in this model resulted from decreased cell osteosarcoma cell proliferation, supporting a role of ErbB3 in bone tumor growth in vivo. Taken together, the results reveal that ErbB3 expression in human osteosarcoma correlates with tumor grade. Furthermore, silencing ErbB3 in a murine osteosarcoma model results in decreased cell growth and invasiveness in vitro, and reduced tumor growth in vivo, which supports the potential therapeutic interest of targeting ErbB3 in osteosarcoma.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/patología , Osteosarcoma/patología , Receptor ErbB-3/genética , Animales , Secuencia de Bases , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Osteosarcoma/genética , Osteosarcoma/metabolismo , ARN Interferente Pequeño , Receptor ErbB-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Stem Cells ; 31(7): 1340-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23533197

RESUMEN

The identification of the molecular mechanisms controlling the degradation of regulatory proteins in mesenchymal stromal cells (MSC) may provide clues to promote MSC osteogenic differentiation and bone regeneration. Ubiquitin ligase-dependent degradation of proteins is an important process governing cell fate. In this study, we investigated the role of the E3 ubiquitin ligase c-Cbl in MSC osteoblast differentiation and identified the mechanisms involved in this effect. Using distinct shRNA targeting c-Cbl, we showed that c-Cbl silencing promotes osteoblast differentiation in murine and human MSC, as demonstrated by increased alkaline phosphatase activity, expression of phenotypic osteoblast marker genes (RUNX2, ALP, type 1 collagen), and matrix mineralization in vitro. Coimmunoprecipitation analyses showed that c-Cbl interacts with the transcription factor STAT5, and that STAT5 forms a complex with RUNX2, a master transcription factor controlling osteoblastogenesis. Silencing c-Cbl decreased c-Cbl-mediated STAT5 ubiquitination, increased STAT5 protein level and phosphorylation, and enhanced STAT5 and RUNX2 transcriptional activity. The expression of insulin like growth factor-1 (IGF-1), a target gene of STAT5, was increased by c-Cbl silencing in MSC and in bone marrow stromal cells isolated from c-Cbl deficient mice, suggesting that IGF-1 contributes to osteoblast differentiation induced by c-Cbl silencing in MSC. Consistent with these findings, pharmacological inhibition of STAT5 activity, or neutralization of IGF-1 activity, abrogated the positive effect of c-Cbl knockdown on MSC osteogenic differentiation. Taken together, the data provide a novel functional mechanism by which the ubiquitin ligase c-Cbl regulates the osteoblastic differentiation program in mesenchymal cells by controlling Cbl-mediated STAT5 degradation and activity.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Factor de Transcripción STAT5/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factor de Transcripción STAT5/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética
5.
PLoS One ; 8(1): e55034, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383046

RESUMEN

BACKGROUND: The molecular mechanisms that are involved in the growth and invasiveness of osteosarcoma, an aggressive and invasive primary bone tumor, are not fully understood. The transcriptional co-factor FHL2 (four and a half LIM domains protein 2) acts as an oncoprotein or as a tumor suppressor depending on the tissue context. In this study, we investigated the role of FHL2 in tumorigenesis in osteosarcoma model. METHODOLOGY/PRINCIPAL FINDINGS: Western blot analyses showed that FHL2 is expressed above normal in most human and murine osteosarcoma cells. Tissue microarray analysis revealed that FHL2 protein expression is high in human osteosarcoma and correlates with osteosarcoma aggressiveness. In murine osteosarcoma cells, FHL2 silencing using shRNA decreased canonical Wnt/ß-catenin signaling and reduced the expression of Wnt responsive genes as well as of the key Wnt molecules Wnt5a and Wnt10b. This effect resulted in inhibition of osteosarcoma cell proliferation, invasion and migration in vitro. Using xenograft experiments, we showed that FHL2 silencing markedly reduced tumor growth and lung metastasis occurence in mice. The anti-oncogenic effect of FHL2 silencing in vivo was associated with reduced cell proliferation and decreased Wnt signaling in the tumors. CONCLUSION/SIGNIFICANCE: Our findings demonstrate that FHL2 acts as an oncogene in osteosarcoma cells and contributes to tumorigenesis through Wnt signaling. More importantly, FHL2 depletion greatly reduces tumor cell growth and metastasis, which raises the potential therapeutic interest of targeting FHL2 to efficiently impact primary bone tumors.


Asunto(s)
Transformación Celular Neoplásica/genética , Silenciador del Gen , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Osteosarcoma/patología , Transducción de Señal/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Wnt/metabolismo , Animales , Apoptosis/genética , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/secundario , Ratones , Terapia Molecular Dirigida , Invasividad Neoplásica , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , ARN Interferente Pequeño/genética , beta Catenina/metabolismo
6.
Bone ; 53(1): 6-12, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23201222

RESUMEN

Wnt signaling is an important pathway that controls the osteogenic differentiation of mesenchymal stromal cells (MSC). We previously showed that FHL2, a LIM-only protein with four and a half LIM domains, controls MSC osteogenic differentiation via the canonical Wnt/ß-catenin signaling. In this study, we investigated the role of Wnt proteins in the regulation of MSC differentiation by FHL2. We found that Wnt3a increased FHL2 mRNA expression in murine C3H10T1/2 mesenchymal cells. Silencing FHL2 using short hairpin (sh) RNA attenuated ß-catenin transcriptional activity and osteogenic differentiation induced by Wnt3a. In addition, FHL2 silencing reduced the expression of the key molecules Wnt5a and Wnt10b and osteoblast gene expression. Wnt10b overcomes the negative effect of FHL2 knockdown on osteoblast gene expression in vitro. To confirm this finding in vivo, we analyzed the expression of these Wnt molecules in FHL2 deficient mice. Histomorphometric analyses showed that FHL2 knockout decreased trabecular number and thickness and reduced bone mass in 15-month old mice. This phenotype was associated with decreased Wnt5a and Wnt10b and lower than normal c-myc, cyclin D1 and osteoblast gene expression in the bone marrow. Ex vivo analysis showed decreased basal and Wnt3a-induced Wnt5a and Wnt10b mRNA expression in FHL2-deficient bone marrow cells, further indicating that this defect may contribute to the reduced osteoblast function in FHL2 deficient mice. In contrast, the decreased adipogenesis induced by FHL2 deficiency in vitro and in vivo was linked to increased Foxo1 expression. Collectively, the results provide evidence for a previously unrecognized mechanism by which FHL2 controls the osteogenic differentiation of MSC, bone formation and bone mass through modulation of Wnt molecules.


Asunto(s)
Diferenciación Celular , Proteínas con Homeodominio LIM/fisiología , Células Madre Mesenquimatosas/citología , Proteínas Musculares/fisiología , Factores de Transcripción/fisiología , Proteínas Wnt/fisiología , Animales , Apoptosis , Western Blotting , Línea Celular , Proliferación Celular , Silenciador del Gen , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Proteínas Musculares/genética , Osteoblastos/citología , Reacción en Cadena de la Polimerasa , Factores de Transcripción/genética , Proteína Wnt-5a
7.
J Bone Miner Res ; 27(10): 2118-29, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22550000

RESUMEN

Alterations of Wnt signaling appear to be involved in the pathogenesis of osteosarcoma, presenting mutations of adenomatous polyposis coli (APC) and epigenetic downregulation of Wnt inhibitory factor 1. However, the precise role of Wnt effectors in the bone cancer progression remains unclear. We previously showed that Wnt/ß-catenin/T-cell factor (TCF) activation are responsible for the repression of syndecan-2, a key modulator of apoptosis and chemosensitivity in osteosarcoma cells, suggesting a role of Wnt signaling in chemoresistance. In this study, we investigated the functional relationship between syndecan-2, Wnt/ß-catenin/TCF signaling and chemosensitivity in these cells. To this goal, we selected resistant osteosarcoma cells from sensitive human cell lines using repeated exposures to doxorubicin. In doxorubicin-responsive but not in doxorubicin-resistant-derived cells syndecan-2 expression was upregulated by doxorubicin treatment. Moreover, syndecan-2 overexpression restored the sensitivity to doxorubicin in resistant-derived cells. We found that syndecan-2 induction by doxorubicin is forkhead box protein O3A (Foxo3a)-dependent. Foxo3a overexpression resulted in increased syndecan-2 expression in sensitive and resistant-derived cells. Doxorubicin modulated Foxo3a binding on syndecan-2 gene promoter and induced Foxo-dependent inhibition of Wnt/TCF activity. Conversely, ß-catenin/TCF activation impaired syndecan-2 induction by doxorubicin, indicating that Wnt signaling is competing with the action of the cytotoxic drug. However, ß-catenin was also found to be required for Foxo3a activity. Consistently, Dickkopf 1 (DKK1) and secreted frizzled-related protein 1 (sFRP-1) altered doxorubicin action in sensitive cells, whereas inhibition of TCF activity strongly decreased cell viability and increased sensitivity to doxorubicin in sensitive and resistant cells. TCF inhibition also increased the effect of doxorubicin treatment in an orthotopic bone tumor model in mice. Altogether, these data provide evidence that the repression of syndecan-2 by Wnt/ß-catenin/TCF signaling contributes to the resistance of osteosarcoma cells to doxorubicin and suggest that TCF inhibition may represent a novel therapeutic strategy in osteosarcoma.


Asunto(s)
Neoplasias Óseas/patología , Doxorrubicina/uso terapéutico , Marcación de Gen , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Sindecano-2/metabolismo , Factores de Transcripción TCF/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción TCF/genética , Transcripción Genética/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
8.
J Bone Miner Res ; 27(10): 2108-17, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22623369

RESUMEN

Targeting receptor tyrosine kinase (RTK) degradation may be an interesting approach to reduce RTK cell signaling in cancer cells. Here we show that increasing E3 ubiquitin ligase casitas B-lineage lymphoma (c-Cbl) expression using lentiviral infection decreased osteosarcoma cell replication and survival and reduced cell migration and invasion in murine and human osteosarcoma cells. Conversely, c-Cbl inhibition using short hairpin RNA (shRNA) increased osteosarcoma cell growth and survival, as well as invasion and migration, indicating that c-Cbl plays a critical role as a bone tumor suppressor. Importantly, the anticancer effect of increasing c-Cbl expression in osteosarcoma cells was related mainly to the downregulation of epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor alpha (PDGFRα). In a murine bone tumor model, increasing c-Cbl expression also reduced RTK expression, resulting in decreased tumor cell proliferation and survival and reduced tumor growth. Interestingly, increasing c-Cbl also markedly reduced lung metastasis in mice. Tissue microarray analysis revealed that low c-Cbl protein expression is associated with elevated EGFR and PDGFRα protein levels in human osteosarcoma with poor outcome. This study shows that increasing c-Cbl expression reduces osteosarcoma cell growth, survival, and metastasis in part through downregulation of RTKs, which supports the potential therapeutic interest of targeting c-Cbl in malignant bone diseases involving increased RTK.


Asunto(s)
Transformación Celular Neoplásica/patología , Marcación de Gen , Osteosarcoma/enzimología , Osteosarcoma/patología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Osteoblastos/enzimología , Osteoblastos/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo
9.
Int J Cancer ; 130(11): 2514-25, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21681744

RESUMEN

Bone tumors strongly influence normal tissues and stimulate bone cells for the production of cytokines supporting proliferation and abnormal survival in cancer cells. We previously reported that the proteoglycan syndecan-2 controls the activity of various cytokines and growth factors and also modulates apoptosis and response to cytotoxic agents in osteosarcoma cell lines. Here, we show that syndecan-2 has a stronger tumor suppressor activity in vivo. We identify calpain-6 as a target gene downregulated by syndecan-2 in cells and in vivo. We demonstrate that calpain-6 expression in osteosarcoma cells depends on endothelin-1, a mediator of the tumor progression in bone. Syndecan-2 overexpression alters ERK1/2, PI3K/AKT and NFκB pathways that are calpain-6-promoting signals downstream of endothelin-1. Immunohistochemical analysis shows that calpain-6 is expressed in human bone tumors and metastases. A high expression of calpain-6 was specially found in recurrent osteosarcoma. Moreover, calpain-6 levels in primary tumors were inversely related to the response to chemotherapy. Consistently, calpain-6 was increased by doxorubicin and was found to be expressed at higher levels in doxorubicin-resistant U2OS osteosarcoma-derived cells as compared to responsive cells. Inhibition of calpain-6 with shRNA resulted in decreased proliferation, increased spontaneous apoptosis and increased sensitivity to doxorubicin and also methotrexate in responsive and resistant osteosarcoma cells. Taken together, our data show that syndecan-2 exerts its pro-apoptotic function through modulation of the endothelin-1/NFκB signaling and through downregulation of calpain-6, a protective factor that contributes to abnormal cell survival. Thus, this study identifies calpain-6 as a new possible therapeutic target in chemoresistant osteosarcoma.


Asunto(s)
Neoplasias Óseas/patología , Calpaína/fisiología , Resistencia a Antineoplásicos , Endotelina-1/fisiología , Osteosarcoma/patología , Transducción de Señal/fisiología , Apoptosis , Neoplasias Óseas/tratamiento farmacológico , Línea Celular Tumoral , Humanos , FN-kappa B/fisiología , Osteosarcoma/tratamiento farmacológico , Sindecano-2/fisiología
10.
Cancer Res ; 70(13): 5399-408, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20530678

RESUMEN

Osteosarcoma is characterized by frequent relapse and metastatic disease associated with resistance to chemotherapy. We previously showed that syndecan-2 is a mediator of the antioncogenic effect of chemotherapeutic drugs. The purpose of this work was to elucidate molecular mechanisms responsible for the low expression of syndecan-2 in osteosarcoma. We compared the regulatory activity of cis-acting DNA sequences of the syndecan-2 gene in osteosarcoma and osteoblastic cell lines. We identified a DNA region that negatively regulates syndecan-2 transcription in the osteosarcoma cells. T-cell factors (TCF) bind to this sequence in vivo. Wnt3a stimulation, beta-catenin activation, and TCF overexpression resulted in syndecan-2 repression, whereas Wnt inhibition using sFRP-1 increased syndecan-2 expression in U2OS cells. RhoA activation blunted the stimulatory effect of sFRP-1 on syndecan-2 transcription, whereas RhoA inhibition enhanced syndecan-2 expression. These results indicate that Wnt/beta-catenin and Wnt/RhoA signaling contribute to syndecan-2 repression. The alteration of syndecan-2 expression in osteosarcoma cell lines also seemed to be related to a higher shedding, controlled by Wnt/RhoA. Conversely, syndecan-2 was found to activate its own expression in U2OS cells through RhoA inhibition. These data identify a molecular network that may contribute to the low expression of the proapoptotic proteoglycan syndecan-2 in osteosarcoma cells. The high activity of the canonical Wnt pathway in the different osteosarcoma cells induces a constitutive repression of syndecan-2 transcription, whereas Wnt/RhoA signaling blocks the amplification loop of syndecan-2 expression. Our results identify syndecan-2 as a Wnt target and bring new insights into a possible pathologic role of Wnt signaling in osteosarcoma.


Asunto(s)
Neoplasias Óseas/genética , Osteosarcoma/genética , Sindecano-2/genética , Proteínas Wnt/metabolismo , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Osteosarcoma/metabolismo , Transducción de Señal , Sindecano-2/biosíntesis , Transcripción Genética , Proteínas Wnt/genética , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...