Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Genes Dev ; 28(14): 1578-91, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25030697

RESUMEN

Lineage or cell of origin of cancers is often unknown and thus is not a consideration in therapeutic approaches. Alveolar rhabdomyosarcoma (aRMS) is an aggressive childhood cancer for which the cell of origin remains debated. We used conditional genetic mouse models of aRMS to activate the pathognomonic Pax3:Foxo1 fusion oncogene and inactivate p53 in several stages of prenatal and postnatal muscle development. We reveal that lineage of origin significantly influences tumor histomorphology and sensitivity to targeted therapeutics. Furthermore, we uncovered differential transcriptional regulation of the Pax3:Foxo1 locus by tumor lineage of origin, which led us to identify the histone deacetylase inhibitor entinostat as a pharmacological agent for the potential conversion of Pax3:Foxo1-positive aRMS to a state akin to fusion-negative RMS through direct transcriptional suppression of Pax3:Foxo1.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Piridinas/farmacología , Rabdomiosarcoma Alveolar/patología , Animales , Línea Celular Tumoral , Linaje de la Célula , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
FASEB J ; 23(8): 2681-90, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19332644

RESUMEN

Bioluminescent reporter genes are sensitive in situ tools for following disease progression in preclinical models, albeit they are subject to scattering and absorption in deep tissues. We have generated a bicistronic Cre/LoxP reporter mouse line that pairs the expression of firefly luciferase with quantifiable expression of a human placental alkaline phosphatase that is secreted into the serum (SeAP). With the use of this dual-modality bioreporter with a novel, inducible Pax7-CreER line for tracking muscle satellite cells, we demonstrate the longitudinal kinetics of muscle stem cell turnover, accounting for a doubling of the signal from satellite cell and progeny every 3.93 wk in the transition from adolescence to early adulthood. We also show that this dual-modality bioreporter can be incorporated in preclinical cancer models, whereby SeAP activity is reflective of tumor burden. Thus, this dual bioreporter permits both spatial localization and accurate quantification of biological processes in vivo even when the tissue of interest is deep within the animal.


Asunto(s)
Células Madre Adultas/metabolismo , Genes Reporteros , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Fosfatasa Alcalina/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Proteínas Ligadas a GPI , Humanos , Isoenzimas/genética , Luciferasas de Luciérnaga/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción PAX7/genética
3.
Muscle Nerve ; 42(2): 245-51, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20544935

RESUMEN

The near-infrared wavelengths (700-900 nm) are the most suitable optical window for light penetration and deep tissue imaging in small animals. Herein we report a near-infrared fluorescent contrast agent, crimson carrier, which acts as a blood pool contrast agent to detect and quantify injury and disease in live animals. After determining the excitation-emission spectra and pharmacokinetics, crimson carrier was injected into myoinjured mice to monitor their recovery. Crimson carrier was also used to image transgenic mice with spontaneous tumors. Crimson carrier has maximal excitation and emission wavelengths of 745 nm and 820 nm, respectively. Elimination occurs predominantly via urinary excretion. We demonstrate the utility of this contrast agent for serial imaging of traumatized muscle as well as muscle tumors. The unique long-acting pharmacokinetics and urinary excretion route characteristics make crimson carrier a contrast agent of choice for the visualization of tumors and injured muscle or other tissues in live animal studies.


Asunto(s)
Medios de Contraste/farmacocinética , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Animales , Colorantes Fluorescentes/farmacocinética , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Transgénicos , Enfermedades Musculares/diagnóstico , Neoplasias/diagnóstico , Análisis de Regresión , Espectroscopía Infrarroja Corta/métodos
4.
Pediatr Blood Cancer ; 53(2): 136-44, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19213072

RESUMEN

BACKGROUND: Tumor initiation has been attributed to haploinsufficiency at a single locus for a large number of cancers. Patched1 (Ptc1) was one of the first such loci, and Ptc1 haploinsufficiency has been asserted to lead to medulloblastoma and rhabdomyosarcoma in mice. PROCEDURE: To study the role of Ptc1 in cerebellar tumor development and to create a preclinical therapeutic platform, we have generated a conditional Ptc1 haploinsufficiency model of medulloblastoma by inactivating Ptc1 in Pax7-expressing cells of the cerebellum. RESULTS: These mice developed exclusively medulloblastoma. We show that despite the presence of transcription of Ptc1, Ptc1 protein is nearly undetectable or absent in tumors. Our results suggest that Ptc1 loss of function is complete, but achieved at the protein level rather than by the classic genetic two-hit mechanism or a strict half-dosage genetic haploinsufficiency mechanism. Furthermore, we found that bortezomib, a 26S proteasome inhibitor, had a significant anti-tumor activity in vitro and in vivo, which was accompanied by restoration of Ptc1 protein and downregulation of the hedgehog signaling pathway. The same effect was seen for both human and mouse medulloblastoma tumor cell growth. CONCLUSIONS: These results suggest that proteasome inhibition is a potential new therapeutic approach in medulloblastoma.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Neoplasias Cerebelosas/genética , Meduloblastoma/genética , Procesamiento Proteico-Postraduccional/fisiología , Pirazinas/farmacología , Receptores de Superficie Celular/efectos de los fármacos , Animales , Bortezomib , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Pérdida de Heterocigocidad/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Ratones , Ratones Noqueados , Factor de Transcripción PAX7 , Receptores Patched , Receptor Patched-1 , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
5.
Mol Cancer Ther ; 10(4): 697-707, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21447712

RESUMEN

Inhibition of the insulin-like growth factor 1 receptor (Igf1r) is an approach being taken in clinical trials to overcome the dismal outcome for metastatic alveolar rhabdomyosarcoma (ARMS), an aggressive muscle cancer of children and young adults. In our study, we address the potential mechanism(s) of Igf1r inhibitor resistance that might be anticipated for patients. Using a genetically engineered mouse model of ARMS, validated for active Igf1r signaling, we show that the prototypic Igf1r inhibitor NVP-AEW541 can inhibit cell growth and induce apoptosis in vitro in association with decreased Akt and Mapk phosphorylation. However, drug resistance in vivo is more common and is accompanied by Igf1r overexpression, Mapk reactivation, and Her2 overexpression. Her2 is found to form heterodimers with Igf1r in resistant primary tumor cell cultures, and stimulation with Igf2 leads to Her2 phosphorylation. The Her2 inhibitor lapatinib cooperates with NVP-AEW541 to reduce Igf1r phosphorylation and to inhibit cell growth even though lapatinib alone has little effect on growth. These results point to the potential therapeutic importance of simultaneous targeting of Igf1r and Her2 to abrogate resistance.


Asunto(s)
Pirimidinas/farmacología , Pirroles/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Rabdomiosarcoma/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Niño , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/patología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Lapatinib , Ratones , Fosforilación/efectos de los fármacos , Codorniz , Quinazolinas/farmacología , Interferencia de ARN , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Adulto Joven
6.
Cancer Cell ; 19(2): 177-91, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21316601

RESUMEN

Embryonal rhabdomyosarcoma (eRMS) shows the most myodifferentiation among sarcomas, yet the precise cell of origin remains undefined. Using Ptch1, p53 and/or Rb1 conditional mouse models and controlling prenatal or postnatal myogenic cell of origin, we demonstrate that eRMS and undifferentiated pleomorphic sarcoma (UPS) lie in a continuum, with satellite cells predisposed to giving rise to UPS. Conversely, p53 loss in maturing myoblasts gives rise to eRMS, which have the highest myodifferentiation potential. Regardless of origin, Rb1 loss modifies tumor phenotype to mimic UPS. In human sarcomas that lack pathognomic chromosomal translocations, p53 loss of function is prevalent, whereas Shh or Rb1 alterations likely act primarily as modifiers. Thus, sarcoma phenotype is strongly influenced by cell of origin and mutational profile.


Asunto(s)
Rabdomiosarcoma Embrionario/patología , Sarcoma/patología , Animales , Diferenciación Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes de Retinoblastoma , Genes p53 , Humanos , Ratones , Mutación , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Rabdomiosarcoma Embrionario/genética , Sarcoma/genética
7.
Mol Cancer Ther ; 9(8): 2354-64, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20663932

RESUMEN

Genetically engineered mouse models (GEMM) of cancer are of increasing value to preclinical therapeutics. Optical imaging is a cost-effective method of assessing deep-seated tumor growth in GEMMs whose tumors can be encoded to express luminescent or fluorescent reporters, although reporter signal attenuation would be improved if animals were fur-free. In this study, we sought to determine whether hereditable furlessness resulting from a hypomorphic mutation in the Hairless gene would or would not also affect immune competence. By assessing humoral and cellular immunity of the SKH1 mouse line bearing the hypomorphic Hairless mutation, we determined that blood counts, immunoglobulin levels, and CD4+ and CD8+ T cells were comparable between SKH1 and the C57Bl/6 strain. On examination of T-cell subsets, statistically significant differences in naïve T cells (1.7 versus 3.4 x 10(5) cells/spleen in SKH1 versus C57Bl/6, P = 0.008) and memory T cells (1.4 versus 0.13 x 10(6) cells/spleen in SKH1 versus C57Bl/6, P = 0.008) were detected. However, the numerical differences did not result in altered T-cell functional response to antigen rechallenge (keyhole limpet hemocyanin) in a lymph node cell in vitro proliferative assay. Furthermore, interbreeding the SKH1 mouse line to a rhabdomyosarcoma GEMM showed preserved antitumor responses of CD56+ natural killer cells and CD163+ macrophages, without any differences in tumor pathology. The fur-free GEMM was also especially amenable to multiplex optical imaging. Thus, SKH1 represents an immune competent, fur-free mouse strain that may be of use for interbreeding to other genetically engineered mouse models of cancer for improved preclinical studies.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ingeniería Genética , Inmunocompetencia/inmunología , Alopecia/inmunología , Alopecia/patología , Animales , Recuento de Células Sanguíneas , Modelos Animales de Enfermedad , Femenino , Imagenología Tridimensional , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inmunización , Subgrupos Linfocitarios/inmunología , Masculino , Ratones , Ratones Pelados , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T/inmunología
8.
Cancer Res ; 69(7): 2902-11, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339268

RESUMEN

The highly aggressive muscle cancer alveolar rhabdomyosarcoma (ARMS) is one of the most common soft tissue sarcoma of childhood, yet the outcome for the unresectable and metastatic disease is dismal and unchanged for nearly three decades. To better understand the pathogenesis of this disease and to facilitate novel preclinical approaches, we previously developed a conditional mouse model of ARMS by faithfully recapitulating the genetic mutations observed in the human disease, i.e., activation of Pax3:Fkhr fusion gene with either p53 or Cdkn2a inactivation. In this report, we show that this model recapitulates the immunohistochemical profile and the rapid progression of the human disease. We show that Pax3:Fkhr expression increases during late preneoplasia but tumor cells undergoing metastasis are under apparent selection for Pax3:Fkhr expression. At a whole-genome level, a cross-species gene set enrichment analysis and metagene projection study showed that our mouse model is most similar to human ARMS when compared with other pediatric cancers. We have defined an expression profile conserved between mouse and human ARMS, as well as a Pax3:Fkhr signature, including the target gene, SKP2. We further identified 7 "druggable" kinases overexpressed across species. The data affirm the accuracy of this genetically engineered mouse model.


Asunto(s)
Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/patología , Alelos , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Progresión de la Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/biosíntesis , Factores de Transcripción Paired Box/genética , Penetrancia , Rabdomiosarcoma Alveolar/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA