Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 42(1): 120, 2023 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-37170152

RESUMEN

BACKGROUND: MET-driven acquired resistance is emerging with unanticipated frequency in patients relapsing upon molecular therapy treatments. However, the determination of MET amplification remains challenging using both standard and next-generation sequencing-based methodologies. Liquid biopsy is an effective, non-invasive approach to define cancer genomic profiles, track tumor evolution over time, monitor treatment response and detect molecular resistance in advance. Circular RNAs (circRNAs), a family of RNA molecules that originate from a process of back-splicing, are attracting growing interest as potential novel biomarkers for their stability in body fluids. METHODS: We identified a circRNA encoded by the MET gene (circMET) and exploited blood-derived cell-free RNA (cfRNA) and matched tumor tissues to identify, stratify and monitor advanced cancer patients molecularly characterized by high MET activity, generally associated with genomic amplification. RESULTS: Using publicly available bioinformatic tools, we discovered that the MET locus transcribes several circRNA molecules, but only one candidate, circMET, was particularly abundant. Deeper molecular analysis revealed that circMET levels positively correlated with MET expression and activity, especially in MET-amplified cells. We developed a circMET-detection strategy and, in parallel, we performed standard FISH and IHC analyses in the same specimens to assess whether circMET quantification could identify patients displaying high MET activity. Longitudinal monitoring of circMET levels in the plasma of selected patients revealed the early emergence of MET amplification as a mechanism of acquired resistance to molecular therapies. CONCLUSIONS: We found that measurement of circMET levels allows identification and tracking of patients characterized by high MET activity. Circulating circMET (ccMET) detection and analysis could be a simple, cost-effective, non-invasive approach to better implement patient stratification based on MET expression, as well as to dynamically monitor over time both therapy response and clonal evolution during treatment.


Asunto(s)
Neoplasias , ARN Circular , Humanos , Biomarcadores , Biología Computacional , Neoplasias/genética , ARN/genética , ARN/metabolismo , ARN Circular/genética
2.
Cancer Res ; 76(20): 6095-6106, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27569217

RESUMEN

Current therapeutic options for the pediatric cancer rhabdomyosarcoma have not improved significantly, especially for metastatic rhabdomyosarcoma. In the current work, we performed a deep miRNA profiling of the three major human rhabdomyosarcoma subtypes, along with cell lines and normal muscle, to identify novel molecular circuits with therapeutic potential. The signature we determined could discriminate rhabdomyosarcoma from muscle, revealing a subset of muscle-enriched miRNA (myomiR), including miR-22, which was strongly underexpressed in tumors. miR-22 was physiologically induced during normal myogenic differentiation and was transcriptionally regulated by MyoD, confirming its identity as a myomiR. Once introduced into rhabdomyosarcoma cells, miR-22 decreased cell proliferation, anchorage-independent growth, invasiveness, and promoted apoptosis. Moreover, restoring miR-22 expression blocked tumor growth and prevented tumor dissemination in vivo Gene expression profiling analysis of miR-22-expressing cells suggested TACC1 and RAB5B as possible direct miR-22 targets. Accordingly, loss- and gain-of-function experiments defined the biological relevance of these genes in rhabdomyosarcoma pathogenesis. Finally, we demonstrated the ability of miR-22 to intercept and overcome the intrinsic resistance to MEK inhibition based on ERBB3 upregulation. Overall, our results identified a novel miR-22 regulatory network with critical therapeutic implications in rhabdomyosarcoma. Cancer Res; 76(20); 6095-106. ©2016 AACR.


Asunto(s)
Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , MicroARNs/fisiología , Rabdomiosarcoma/terapia , Animales , Diferenciación Celular , Línea Celular Tumoral , Femenino , Proteínas Fetales/genética , Proteínas Fetales/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteína MioD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Receptor ErbB-3/genética , Receptor ErbB-3/fisiología , Rabdomiosarcoma/etiología , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/fisiología
3.
Elife ; 52016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26987019

RESUMEN

Embryonal Rhabdomyosarcoma (ERMS) and Undifferentiated Pleomorphic Sarcoma (UPS) are distinct sarcoma subtypes. Here we investigate the relevance of the satellite cell (SC) niche in sarcoma development by using Hepatocyte Growth Factor (HGF) to perturb the niche microenvironment. In a Pax7 wild type background, HGF stimulation mainly causes ERMS that originate from satellite cells following a process of multistep progression. Conversely, in a Pax7 null genotype ERMS incidence drops, while UPS becomes the most frequent subtype. Murine EfRMS display genetic heterogeneity similar to their human counterpart. Altogether, our data demonstrate that selective perturbation of the SC niche results in distinct sarcoma subtypes in a Pax7 lineage-dependent manner, and define a critical role for the Met axis in sarcoma initiation. Finally, our results provide a rationale for the use of combination therapy, tailored on specific amplifications and activated signaling pathways, to minimize resistance emerging from sarcomas heterogeneity.


Asunto(s)
Proliferación Celular , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Transcripción PAX7/metabolismo , Sarcoma/patología , Animales , Humanos , Ratones Transgénicos , Factor de Transcripción PAX7/genética , Sarcoma/genética
4.
Cell Cycle ; 14(9): 1389-402, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25644430

RESUMEN

Rhadomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. RMS cells resemble fetal myoblasts but are unable to complete myogenic differentiation. In previous work we showed that miR-206, which is low in RMS, when induced in RMS cells promotes the resumption of differentiation by modulating more than 700 genes. To better define the pathways involved in the conversion of RMS cells into their differentiated counterpart, we focused on 2 miR-206 effectors emerged from the microarray analysis, SMYD1 and G6PD. SMYD1, one of the most highly upregulated genes, is a H3K4 histone methyltransferase. Here we show that SMYD1 silencing does not interfere with the proliferative block or with the loss anchorage independence imposed by miR-206, but severely impairs differentiation of ERMS, ARMS, and myogenic cells. Thus SMYD1 is essential for the activation of muscle genes. Conversely, among the downregulated genes, we found G6PD, the enzyme catalyzing the rate-limiting step of the pentose phosphate shunt. In this work, we confirmed that G6PD is a direct target of miR-206. Moreover, we showed that G6PD silencing in ERMS cells impairs proliferation and soft agar growth. However, G6PD overexpression does not interfere with the pro-differentiating effect of miR-206, suggesting that G6PD downmodulation contributes to - but is not an absolute requirement for - the tumor suppressive potential of miR-206. Targeting cancer metabolism may enhance differentiation. However, therapeutic inhibition of G6PD is encumbered by side effects. As an alternative, we used DCA in combination with miR-206 to increase the flux of pyruvate into the mitochondrion by reactivating PDH. DCA enhanced the inhibition of RMS cell growth induced by miR-206, and sustained it upon miR-206 de-induction. Altogether these results link miR-206 to epigenetic and metabolic reprogramming, and suggest that it may be worth combining differentiation-inducing with metabolism-directed approaches.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , MicroARNs/metabolismo , Desarrollo de Músculos , Proteínas Musculares/metabolismo , Rabdomiosarcoma Alveolar/enzimología , Rabdomiosarcoma Embrionario/enzimología , Factores de Transcripción/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/genética , Ácido Dicloroacético/farmacología , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Glucosafosfato Deshidrogenasa/genética , Humanos , MicroARNs/genética , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/genética , Mioblastos/enzimología , Mioblastos/patología , Fenotipo , Interferencia de ARN , Rabdomiosarcoma Alveolar/tratamiento farmacológico , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/patología , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Transfección
5.
PLoS One ; 6(2): e14675, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21347410

RESUMEN

BACKGROUND: The Hepatocyte Growth Factor (HGF) is a pleiotropic cytokine involved in many physiological processes, including skeletal muscle, placenta and liver development. Little is known about its role and that of Met tyrosine kinase receptor in cardiac development. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we generated two transgenic mice with cardiac-specific, tetracycline-suppressible expression of either Hepatocyte Growth Factor (HGF) or the constitutively activated Tpr-Met kinase to explore: i) the effect of stimulation of the endogenous Met receptor by autocrine production of HGF and ii) the consequence of sustained activation of Met signalling in the heart. We first showed that Met is present in the neonatal cardiomyocytes and is responsive to exogenous HGF. Exogenous HGF starting from prenatal stage enhanced cardiac proliferation and reduced sarcomeric proteins and Connexin43 (Cx43) in newborn mice. As adults, these transgenics developed systolic contractile dysfunction. Conversely, prenatal Tpr-Met expression was lethal after birth. Inducing Tpr-Met expression during postnatal life caused early-onset heart failure, characterized by decreased Cx43, upregulation of fetal genes and hypertrophy. CONCLUSIONS/SIGNIFICANCE: Taken together, our data show that excessive activation of the HGF/Met system in development may result in cardiac damage and suggest that Met signalling may be implicated in the pathogenesis of cardiac disease.


Asunto(s)
Cardiopatías/metabolismo , Corazón/crecimiento & desarrollo , Miocardio/enzimología , Miocardio/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Conexina 43/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/fisiopatología , Cardiopatías/enzimología , Cardiopatías/etiología , Cardiopatías/patología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/farmacología , Ratones , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Especificidad de Órganos , Transducción de Señal/efectos de los fármacos
6.
Cell Cycle ; 9(5): 918-22, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20160508

RESUMEN

Differentiation involves repression of genes governing proliferation and self-renewal, and transcriptional activation of lineage-specific genes. The mechanisms underlying these changes are epigenetic. In cancer cells differentiation genes are locked into a transcriptionally inactive state. Recent results show that in spite of the diversity of the genetic lesions leading to a cancerous phenotype, it may still be possible to release this block and to force differentiation. The key may be microRNAs (miRNAs) which directly or indirectly target epigenetic modifiers. These miRNAs could allow to apply to solid tumors the non-toxic differentiative approach currently adopted in some haematologic malignancies.


Asunto(s)
MicroARNs/metabolismo , Neoplasias/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Epigénesis Genética , Ratones , Neoplasias/metabolismo
7.
J Clin Invest ; 119(8): 2366-78, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19620785

RESUMEN

Many microRNAs (miRNAs), posttranscriptional regulators of numerous cellular processes and developmental events, are downregulated in tumors. However, their role in tumorigenesis remains largely unknown. In this work, we examined the role of the muscle-specific miRNAs miR-1 and miR-206 in human rhabdomyosarcoma (RMS), a soft tissue sarcoma thought to arise from skeletal muscle progenitors. We have shown that miR-1 was barely detectable in primary RMS of both the embryonal and alveolar subtypes and that both miR-1 and miR-206 failed to be induced in RMS cell lines upon serum deprivation. Moreover, reexpression of miR-206 in RMS cells promoted myogenic differentiation and blocked tumor growth in xenografted mice by switching the global mRNA expression profile to one that resembled mature muscle. Finally, we showed that the product of the MET proto-oncogene, the Met tyrosine-kinase receptor, which is overexpressed in RMS and has been implicated in RMS pathogenesis, was downregulated in murine satellite cells by miR-206 at the onset of normal myogenesis. Thus, failure of posttranscriptional modulation may underlie Met overexpression in RMS and other types of cancer. We propose that tissue-specific miRNAs such as miR-1 and miR-206, given their ability to modulate hundreds of transcripts and to act as nontoxic differentiating agents, may override the genomic heterogeneity of solid tumors and ultimately hold greater therapeutic potential than single gene-directed drugs.


Asunto(s)
MicroARNs/fisiología , Desarrollo de Músculos/fisiología , Rabdomiosarcoma/prevención & control , Animales , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Humanos , Ratones , MicroARNs/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-met , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/fisiología , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 14(7): 2220-6, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381964

RESUMEN

PURPOSE: Met, the tyrosine kinase receptor for hepatocyte growth factor, is frequently deregulated in human cancer. Recent evidence indicates that Met amplification may confer resistance to treatments directed toward other receptor tyrosine kinases. Thus, there is a need to develop Met inhibitors into therapeutic tools, to be used alone or in combination with other molecularly targeted drugs. Preclinical validation of Met inhibitors has thus far been done in nude mice bearing cancer cells xenografts. A far superior model would be a transgenic line developing spontaneous Met-driven tumors with high penetrance and short latency. EXPERIMENTAL DESIGN: To this end, we introduced into the mouse genome TPR-MET, the oncogenic form of MET. The Tpr-Met protein ensures deregulation of Met signaling because dimerization motifs in the Tpr moiety cause ligand-independent activation of the Met kinase. RESULTS: Here, we describe a TPR-MET transgenic line that develops thymic T-cell lymphoma with full penetrance and very short latency. In the tumors, Tpr-Met and its effectors were phosphorylated. Treatment of tumor-derived T lymphocytes with the selective Met inhibitor PHA-665752 at nanomolar concentrations abolished phosphorylation of Met and downstream effectors and led to caspase-mediated apoptosis. I.v. administration of PHA-665752 to transgenic mice bearing lymphomas in exponential growth phase led to a significant decrease in tumor growth and, in some cases, to tumor regression. CONCLUSIONS: Our transgenic line, which within 2 months reliably develops Tpr-Met-driven T-cell lymphoma, represents a valuable tool to explore the efficacy and therapeutic potential of Met kinase inhibitors as anticancer drugs.


Asunto(s)
Modelos Animales de Enfermedad , Linfoma/tratamiento farmacológico , Linfoma/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/genética , Animales , Western Blotting , Técnicas de Transferencia de Gen , Humanos , Inmunohistoquímica , Indoles/farmacología , Linfoma/patología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Sulfonas/farmacología
9.
PLoS One ; 3(3): e1828, 2008 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-18350164

RESUMEN

BACKGROUND: Osteosarcoma (OSA) is lethal when metastatic after chemotherapy and/or surgical treatment. Thus animal models are necessary to study the OSA metastatic spread and to validate novel therapies able to control the systemic disease. We report the development of a syngeneic (Balb/c) murine OSA model, using a cell line derived from a spontaneous murine tumor. METHODOLOGY: The tumorigenic and metastatic ability of OSA cell lines were assayed after orthotopic injection in mice distal femur. Expression profiling was carried out to characterize the parental and metastatic cell lines. Cells from metastases were propagated and engineered to express Luciferase, in order to follow metastases in vivo. PRINCIPAL FINDINGS: Luciferase bioluminescence allowed to monitor the primary tumor growth and revealed the appearance of spontaneous pulmonary metastases. In vivo assays showed that metastasis is a stable property of metastatic OSA cell lines after both propagation in culture and luciferase trasduction. When compared to parental cell line, both unmodified and genetically marked metastatic cells, showed comparable and stable differential expression of the enpp4, pfn2 and prkcd genes, already associated to the metastatic phenotype in human cancer. CONCLUSIONS: This OSA animal model faithfully recapitulates some of the most important features of the human malignancy, such as lung metastatization. Moreover, the non-invasive imaging allows monitoring the tumor progression in living mice. A great asset of this model is the metastatic phenotype, which is a stable property, not modifiable after genetic manipulation.


Asunto(s)
Modelos Animales de Enfermedad , Luciferasas/metabolismo , Neoplasias Pulmonares/secundario , Osteosarcoma/patología , Animales , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C
10.
Eur J Cancer ; 44(6): 876-84, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18342500

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood, divided into two major histological subtypes, embryonal (ERMS) and alveolar (ARMS). To explore the possibility that the proteasome could be a target of therapeutic value in rhabdomyosarcoma, we treated several RMS cell lines with the proteasome inhibitor bortezomib (Velcade or PS-341) at a concentration of 13-26 nM. RMS cells showed high sensitivity to the drug, whereas no toxic effect was observed in primary human myoblasts. In both ERMS and ARMS cells bortezomib promoted apoptosis, activation of caspase 3 and 7 and induced a dose-dependent reduction of anchorage-independent growth. Furthermore, bortezomib induced activation of the stress response, cell cycle arrest and the reduction of NF-kappaB transcriptional activity. Finally, bortezomib decreased tumour growth and impaired cells viability, proliferation and angiogenesis in a xenograft model of RMS. In conclusion, our data indicate that bortezomib could represent a novel drug against RMS tumours.


Asunto(s)
Ácidos Borónicos/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteasoma , Pirazinas/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Bortezomib , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/prevención & control , Trasplante Heterólogo , Células Tumorales Cultivadas
11.
Exp Cell Res ; 313(11): 2308-17, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17490646

RESUMEN

PAX3-FKHR, the product of a rearrangement of PAX3 with FKHR is the pathogenetic marker for alveolar rhabdomyosarcoma, an aggressive form of childhood cancer. In this work we show that PAX3-FKHR, which is a stronger transcriptional activator relative to PAX3, can lead to two apparently irreconcilable outcomes: transformation and terminal myogenic differentiation. Fibroblasts (10T1/2, NIH3T3, and a newly established murine line named 'Plus') transduced by PAX3-FKHR acquire transformed features such as anchorage independence and loss of contact inhibition and concomitantly undergo various degrees of myogenic conversion depending on the host cells, including, in the case of the Plus line, terminal differentiation into contractile myotubes. This work highlights the potential of PAX3-FKHR to functionally operate as a deregulated Pangene and may have implications with regard to the identity of the precursor cell giving rise to alveolar rhabdomyosarcoma.


Asunto(s)
Transformación Celular Neoplásica/genética , Fibroblastos/metabolismo , Fibras Musculares Esqueléticas/citología , Proteínas de Fusión Oncogénica/fisiología , Factores de Transcripción Paired Box/fisiología , Animales , Diferenciación Celular , Línea Celular , Humanos , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción Paired Box/genética , Transfección
12.
Mol Biol Cell ; 18(3): 986-94, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17202410

RESUMEN

Ghrelin is an acylated peptidyl gastric hormone acting on the pituitary and hypothalamus to stimulate appetite, adiposity, and growth hormone release, through activation of growth hormone secretagogue receptor (GHSR)-1a receptor. Moreover, ghrelin features several activities such as inhibition of apoptosis, regulation of differentiation, and stimulation or inhibition of proliferation of several cell types. Ghrelin acylation is absolutely required for both GHSR-1a binding and its central endocrine activities. However, the unacylated ghrelin form, des-acyl ghrelin, which does not bind GHSR-1a and is devoid of any endocrine activity, is far more abundant than ghrelin in plasma, and it shares with ghrelin some of its cellular activities. In here we show that both ghrelin and des-acyl ghrelin stimulate proliferating C2C12 skeletal myoblasts to differentiate and to fuse into multinucleated myotubes in vitro through activation of p38. Consistently, both ghrelin and des-acyl ghrelin inhibit C2C12 proliferation in growth medium. Moreover, the ectopic expression of ghrelin in C2C12 enhances differentiation and fusion of these myoblasts in differentiation medium. Finally, we show that C2C12 cells do not express GHSR-1a, but they do contain a common high-affinity binding site recognized by both acylated and des-acylated ghrelin, suggesting that the described activities on C2C12 are likely mediated by this novel, yet unidentified receptor for both ghrelin forms.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Hormonas Peptídicas/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Biomarcadores , Fusión Celular , Proliferación Celular/efectos de los fármacos , Medios de Cultivo , ADN/biosíntesis , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ghrelina , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ghrelina , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Biol Chem ; 282(9): 6812-22, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17194700

RESUMEN

Skeletal muscle atrophy is a common debilitating feature of many systemic diseases, including cancer. Here we examined the effects of inducing expression of an oncogenic version of the Met receptor (Tpr-Met) in terminally differentiated skeletal muscle. A responder mouse containing the Tpr-Met oncogene and GFP (green fluorescent protein) as a reporter was crossed with a transactivator mouse expressing tTA under the control of the muscle creatine kinase promoter. Tpr-Met induction during fetal development and in young adult mice caused severe muscle wasting, with decreased fiber size and loss of myosin heavy chain protein. Concomitantly, in the Tpr-Met-expressing muscle the mRNA of the E3 ubiquitin ligases atrogin-1/MAFbx, MuRF1, and of the lysosomal protease cathepsin L, which are markers of skeletal muscle atrophy, was significantly increased. In the same muscles phosphorylation of the Met downstream effectors Akt, p38 MAPK, and IkappaBalpha was higher than in normal controls. Induction of Tpr-Met in differentiating satellite cells derived from the double transgenics caused aberrant cell fusion, protein loss, and myotube collapse. Increased phosphorylation of Met downstream effectors was also observed in the Tpr-Met-expressing myotubes cultures. Treatment of these cultures with either a proteasomal or a p38 inhibitor prevented Tpr-Met-mediated myotube breakdown, establishing accelerated protein degradation consequent to inappropriate activation of p38 as the major route for the Tpr-Met-induced muscle phenotype.


Asunto(s)
Atrofia Muscular/etiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-met/fisiología , Células Satélite del Músculo Esquelético/patología
14.
J Neurosci ; 26(37): 9593-602, 2006 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-16971543

RESUMEN

Hydrocephalus is a common and variegated pathology often emerging in newborn children after genotoxic insults during pregnancy (Hicks and D'Amato, 1980). Cre recombinase is known to have possible toxic effects that can compromise normal cell cycle and survival. Here we show, by using three independent nestin Cre transgenic lines, that high levels of Cre recombinase expression into the nucleus of neuronal progenitors can compromise normal brain development. The transgenics analyzed are the nestin Cre Balancer (Bal1) line, expressing the Cre recombinase with a nuclear localization signal, and two nestin CreER(T2) (Cre recombinase fused with a truncated estrogen receptor) mice lines with different levels of expression of a hybrid CreER(T2) recombinase that translocates into the nucleus after tamoxifen treatment. All homozygous Bal1 nestin Cre embryos displayed reduced neuronal proliferation, increased aneuploidy and cell death, as well as defects in ependymal lining and lamination of the cortex, leading to microencephaly and to a form of communicating hydrocephalus. An essentially overlapping phenotype was observed in the two nestin CreER(T2) transgenic lines after tamoxifen mediated-CreER(T2) translocation into the nucleus. Neither tamoxifen-treated wild-type nor nestin CreER(T2) oil-treated control mice displayed these defects. These results indicate that some forms of hydrocephalus may derive from a defect in neuronal precursors proliferation. Furthermore, they underscore the potential risks for developmental studies of high levels of nuclear Cre in neurogenic cells.


Asunto(s)
Encéfalo/anomalías , Hidrocefalia/enzimología , Integrasas/metabolismo , Microcefalia/enzimología , Malformaciones del Sistema Nervioso/enzimología , Células Madre/enzimología , Aneuploidia , Animales , Biomarcadores/metabolismo , Encéfalo/enzimología , Encéfalo/fisiopatología , Muerte Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Epéndimo/anomalías , Epéndimo/metabolismo , Epéndimo/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hidrocefalia/genética , Hidrocefalia/fisiopatología , Integrasas/genética , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Ratones Transgénicos , Microcefalia/genética , Microcefalia/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/fisiopatología , Nestina , Neuronas/enzimología , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología
15.
Cancer Res ; 66(9): 4742-9, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16651427

RESUMEN

Rhabdomyosarcoma (RMS) is a highly malignant soft-tissue tumor of childhood deriving from skeletal muscle cells. RMS can be classified in two major histologic subtypes: embryonal (ERMS) and alveolar (ARMS), the latter being characterized by the PAX3/7-FKHR translocation. Here we first investigated whether the Met receptor, a transcriptional target of PAX3 and PAX7, has a role in PAX3-FKHR-mediated transformation. Following PAX3-FKHR transduction, Met was up-regulated in mouse embryonal fibroblasts (MEF), NIH 3T3 and C2C12 cells, and they all acquired anchorage independence. This property was lost in low serum but addition of hepatocyte growth factor/scatter factor (HGF/SF) rescued soft-agar growth. Genetic proof that Met is necessary for this PAX3-FKHR-mediated effect was obtained by transducing with PAX3-FKHR MEFs derived from Met mutant (Met(D/D)) and wild-type (Met(+/+)) embryos. Only Met(+/+) MEFs acquired anchorage-independent growth whereas PAX3-FKHR-transduced Met(D/D) cells were unable to form colonies in soft agar. To verify if Met had a role in RMS maintenance, we silenced the receptor by transducing ERMS and ARMS cell lines with an inducible lentivirus expressing an anti-Met short hairpin RNA (shRNA). Met down-regulation significantly affected RMS cells proliferation, survival, invasiveness, and anchorage-independent growth. Finally, induction of the Met-directed shRNA promoted a dramatic reduction of tumor mass in a xenograft model of RMS. Our data show that both ARMS- and ERMS-derived cell lines, in spite of the genetic drift which may have occurred in years of culture, seem to have retained an "addiction" to the Met oncogene and suggest that Met may represent a target of choice to develop novel therapeutic strategies for ARMS.


Asunto(s)
Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/fisiología , Rabdomiosarcoma Alveolar/terapia , Rabdomiosarcoma Embrionario/terapia , Animales , Apoptosis/genética , Procesos de Crecimiento Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Silenciador del Gen , Células HeLa , Factor de Crecimiento de Hepatocito , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Invasividad Neoplásica , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-met , Interferencia de ARN , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Receptores de Factores de Crecimiento/genética , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/metabolismo , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/metabolismo , Rabdomiosarcoma Embrionario/patología , Transducción Genética , Regulación hacia Arriba
16.
Blood ; 107(2): 689-97, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16189272

RESUMEN

Anaplastic large-cell lymphomas (ALCLs) carry chromosome translocations in which the anaplastic lymphoma kinase (ALK) gene is fused to several partners, most frequently, the NPM1 gene. We have demonstrated that the constitutive activation of ALK fusion proteins results in cellular transformation and lymphoid neoplasia. Herein, we specifically down-regulated ALK protein expression by using small hairpin RNA (shRNA) targeting a sequence coding for the catalytic domain of ALK. The ablation of ALK leads to the down-modulation of known ALK downstream effectors, cell growth arrest, and reversion of the transformed phenotype of ALK(+) mouse embryonic fibroblasts in vitro and in vivo. In human ALCL cells lentiviral-mediated ALK knock-down leads to G(1) cell-cycle arrest and apoptosis in vitro and tumor growth inhibition and regression in vivo. Using a specific approach we have demonstrated that the survival and growth of ALK(+) ALCLs are strictly dependent on ALK activation and signaling. Therefore, ALK is a viable target for therapeutic intervention and its inactivation might represent a pivotal approach for the treatment of ALK lymphomas and other ALK-dependent human tumors.


Asunto(s)
Apoptosis , Linfoma Anaplásico de Células Grandes/enzimología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , Quinasa de Linfoma Anaplásico , Animales , Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica , Fibroblastos , Técnica del Anticuerpo Fluorescente , Humanos , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/patología , Ratones , Ratones Desnudos , Nucleofosmina , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Retroviridae/genética , Transfección
17.
Cancer Gene Ther ; 12(5): 456-63, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15719029

RESUMEN

Tpr-Met, the oncogenic counterpart of the Met receptor, has been detected in gastric cancers, as well as in precursor lesions and in the adjacent normal gastric mucosa. This has prompted the suggestion that Tpr-Met may predispose to the development of gastric tumors. Given the sequence specificity of RNA interference, oncogenes activated by point mutation or rearrangements can be targeted while spearing the product of the wild-type allele. In this work, we report specific suppression of Tpr-Met expression and inhibition of Tpr-Met-mediated transformation and tumorigenesis by means of a short interfering RNA (siRNA) directed toward the Tpr-Met junction (anti-TM2). When delivered by a lentiviral vector, anti-TM2 siRNA was effective also in mouse embryonal fibroblasts or epithelial cells expressing high levels of Tpr-Met. Our results suggest that lentiviral-mediated delivery of anti-TM2 siRNA may be developed into a powerful tool to treat Tpr-Met-positive cancers.


Asunto(s)
Vectores Genéticos/genética , Lentivirus/genética , Neoplasias Experimentales/terapia , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Interferencia de ARN , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Terapia Genética , Humanos , Ratones , Neoplasias Experimentales/etiología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , ARN Interferente Pequeño/genética , Transducción Genética
18.
Dev Dyn ; 231(3): 582-91, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15376315

RESUMEN

The Pax3 and c-met genes are necessary for the development of tongue, diaphragm, and limb muscles. These hypaxial muscles derive from precursors that migrate out of the ventrolateral lip of the somites at occipital, cervical, and limb levels. In this work, we re-examined primary myogenesis in c-met signaling mutants using a skeletal muscle-specific lacZ transgene (Mlc3f-nlacZ-2E). This strategy allowed us to identify precisely the shoulder, limb, tongue, and dermal muscles that need Met for development and to confirm that the morphological structure of epaxial and body wall muscles was normal, even in the most severe c-met mutant. Surprisingly, however, X-gal staining showed that, in this mutant, hyoid arch-derived facial muscles were either reduced or absent, thus revealing that Met also contributes to the development of muscles in the head.


Asunto(s)
Músculos Faciales/embriología , Operón Lac , Músculos/embriología , Mutación , Proteínas Proto-Oncogénicas c-met/genética , Animales , Linaje de la Célula , Movimiento Celular , Músculos Faciales/fisiología , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Cinética , Mesodermo/citología , Ratones , Ratones Mutantes , Ratones Transgénicos , Desarrollo de Músculos , Transgenes , beta-Galactosidasa/metabolismo
19.
Genes Dev ; 17(23): 2950-65, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14665670

RESUMEN

Pax3 is a key transcription factor implicated in development and human disease. To dissect the role of Pax3 in myogenesis and establish whether it is a repressor or activator, we generated loss- and gain-of-function alleles by targeting an nLacZ reporter and a sequence encoding the oncogenic fusion protein PAX3-FKHR into the Pax3 locus. Rescue of the Pax3 mutant phenotypes by PAX3-FKHR suggests that Pax3 acts as a transcriptional activator during embryogenesis. This is confirmed by a Pax reporter mouse. However, mice expressing PAX3-FKHR display developmental defects, including ectopic delamination and inappropriate migration of muscle precursor cells. These events result from overexpression of c-met, leading to constitutive activation of Met signaling, despite the absence of the ligand SF/HGF. Haploinsufficiency of c-met rescues this phenotype, confirming the direct genetic link with Pax3. The gain-of-function phenotype is also characterized by overactivation of MyoD. The consequences of PAX3-FKHR myogenic activity in the limbs and cervical and thoracic regions point to differential regulation of muscle growth and patterning. This gain-of-function allele provides a new approach to the molecular and cellular analysis of the role of Pax3 and of its target genes in vivo.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas c-met/fisiología , Transducción de Señal , Transactivadores , Factores de Transcripción/fisiología , Alelos , Animales , Proteínas de Unión al ADN/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Inmunohistoquímica , Ratones , Ratones Mutantes , Proteínas Musculares/fisiología , Proteína MioD/fisiología , Factor 5 Regulador Miogénico , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Fenotipo
20.
Oncogene ; 21(54): 8334-9, 2002 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-12447696

RESUMEN

Activation of tyrosine kinase receptors is associated with human tumors. Tumorigenic versions of several RTKs, such as Ret, Kit and Met carry activating mutations at highly conserved residues of the tyrosine kinase domain. We have investigated the effect of some of these mutations on the NTRK1/NGF receptor, for which no naturally occurring activating point mutations have been so far detected. We introduced the following mutations in NTRK1 tyrosine kinase domain: (i) D668N equivalent to Met D1246N associated to HPRC; (ii) D668V modelled on Kit D816V found in mastocytosis; (iii) M688T corresponding to Ret M918T associated to the cancer syndrome MEN2B. The Met-like mutation rendered the NTRK1 receptor more responsive to ligand, as observed for the corresponding mutation in Met. On the contrary the Kit-like D668V resulted as neutral mutation. Surprisingly, the MEN2B-like M688T completely abrogated NTRK1 receptor activity, resulting as a loss of function mutation. Our results show that the mutations tested, although involving conserved amino acids in highly homologous regions, exert distinct effects in different receptors, and suggest a very peculiar auto-inhibitory mechanism for NTRK1.


Asunto(s)
Mutación , Receptor trkA/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Cartilla de ADN , Datos de Secuencia Molecular , Receptor trkA/química , Receptor trkA/genética , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...