Mostrar el registro sencillo del ítem

dc.contributor.author
Teitz, Tal  
dc.contributor.author
Inoue, Madoka  
dc.contributor.author
Valentine, Marcus B.  
dc.contributor.author
Zhu, Kejin  
dc.contributor.author
Rehg, Jerold E.  
dc.contributor.author
Zhao, Wei  
dc.contributor.author
Finkelstein, David  
dc.contributor.author
Wang, Yong-Dong  
dc.contributor.author
Johnson, Melissa D.  
dc.contributor.author
Calabrese, Christopher  
dc.contributor.author
Rubinstein, Marcelo  
dc.contributor.author
Hakem, Razqallah  
dc.contributor.author
Weiss, William A.  
dc.contributor.author
Lahti, Jill M.  
dc.date.available
2016-02-03T20:30:35Z  
dc.date.issued
2013-03-27  
dc.identifier.citation
Teitz, Tal; Inoue, Madoka; Valentine, Marcus B.; Zhu, Kejin; Rehg, Jerold E.; et al.; Th-MYCN Mice with Caspase-8 Deficiency Develop Advanced Neuroblastoma with Bone Marrow Metastasis; American Association for Cancer Research; Cancer Research; 73; 27-3-2013; 4086-4097  
dc.identifier.issn
0008-5472  
dc.identifier.uri
http://hdl.handle.net/11336/4016  
dc.description.abstract
Neuroblastoma, the most common extracranial pediatric solid tumor, is responsible for 15% of all childhood cancer deaths. Patients frequently present at diagnosis with metastatic disease, particularly to the bone marrow (BM). Advances in therapy and understanding of the metastatic process have been limited due in part, to the lack of animal models harboring BM disease. The widely employed transgenic model, the Th-MYCN mouse, exhibits limited metastasis to this site. Here we establish the first genetic immunocompetent mouse model for metastatic neuroblastoma with enhanced secondary tumors in the BM. This model recapitulates two frequent alterations in metastatic neuroblasoma, over-expression of MYCN and loss of caspase-8 expression. Mouse caspase-8 gene was deleted in neural crest lineage cells by crossing a Th-Cre transgenic mouse with a caspase-8 conditional knockout mouse. This mouse was then crossed with the neuroblastoma prone Th-MYCN mouse. While over-expression of MYCN by itself rarely caused bone marrow metastasis, combining MYCN overexpression and caspase-8 deletion significantly enhanced BM metastasis (37% incidence). Microarray expression studies of the primary tumors mRNAs and microRNAs revealed extracellular matrix (ECM) structural changes, increased expression of genes involved in epithelial to mesenchymal transition, inflammation and down-regulation of miR-7a and miR-29b. These molecular changes have been shown to be associated with tumor progression and activation of the cytokine transforming growth factor beta (TGF-β) pathway in various tumor models. Cytokine TGF-β can preferentially promote single cell motility and blood borne metastasis and therefore activation of this pathway may explain the enhanced BM metastasis observed in this animal model.  
dc.format
application/pdf  
dc.language.iso
eng  
dc.publisher
American Association for Cancer Research  
dc.rights
info:eu-repo/semantics/openAccess  
dc.rights.uri
https://creativecommons.org/licenses/by-nc-sa/2.5/ar/  
dc.subject
Cancer  
dc.subject
Metastasis  
dc.subject
Caspase-8  
dc.subject
Catecholamine  
dc.subject.classification
Oncología  
dc.subject.classification
Medicina Clínica  
dc.subject.classification
CIENCIAS MÉDICAS Y DE LA SALUD  
dc.title
Th-MYCN Mice with Caspase-8 Deficiency Develop Advanced Neuroblastoma with Bone Marrow Metastasis  
dc.type
info:eu-repo/semantics/article  
dc.type
info:ar-repo/semantics/artículo  
dc.type
info:eu-repo/semantics/publishedVersion  
dc.date.updated
2016-03-30 10:35:44.97925-03  
dc.journal.volume
73  
dc.journal.pagination
4086-4097  
dc.journal.pais
Estados Unidos  
dc.journal.ciudad
Philadelphia  
dc.description.fil
Fil: Teitz, Tal. St. Jude Children’s Research Hospital. Department of Tumor Cell Biology; Estados Unidos  
dc.description.fil
Fil: Inoue, Madoka. St. Jude Children’s Research Hospital. Department of Tumor Cell Biology; Estados Unidos  
dc.description.fil
Fil: Valentine, Marcus B.. St. Jude Children’s Research Hospital. Department of Tumor Cell Biology; Estados Unidos  
dc.description.fil
Fil: Zhu, Kejin. St. Jude Children’s Research Hospital. Department of Tumor Cell Biology; Estados Unidos  
dc.description.fil
Fil: Rehg, Jerold E.. St. Jude Children’s Research Hospital. Department of Pathology; Estados Unidos  
dc.description.fil
Fil: Zhao, Wei. St. Jude Children’s Research Hospital. Department of Biostatistics; Estados Unidos  
dc.description.fil
Fil: Finkelstein, David. St. Jude Children’s Research Hospital. Department of Computational Biology; Estados Unidos  
dc.description.fil
Fil: Wang, Yong-Dong. St. Jude Children’s Research Hospital. Hartwell Center for Bioinformatics and Biotechnology; Estados Unidos  
dc.description.fil
Fil: Johnson, Melissa D.. St. Jude Children’s Research Hospital. Animal Imaging Center; Estados Unidos  
dc.description.fil
Fil: Calabrese, Christopher. St. Jude Children’s Research Hospital. Animal Imaging Center; Estados Unidos  
dc.description.fil
Fil: Rubinstein, Marcelo. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Investigaciones en Ingeniería Genética y Biología Molecular; Argentina  
dc.description.fil
Fil: Hakem, Razqallah. University of Toronto. Ontario Cancer Institute. Department of Medical Biophysics; Canadá  
dc.description.fil
Fil: Weiss, William A.. University of California. Departments of Neurology, Pediatrics and Neurological Surgery; Estados Unidos  
dc.description.fil
Fil: Lahti, Jill M.. St. Jude Children’s Research Hospital. Department of Tumor Cell Biology; Estados Unidos  
dc.journal.title
Cancer Research  
dc.relation.alternativeid
info:eu-repo/semantics/altIdentifier/url/http://cancerres.aacrjournals.org/content/73/13/4086.long  
dc.relation.alternativeid
info:eu-repo/semantics/altIdentifier/url/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3702642/  
dc.relation.alternativeid
info:eu-repo/semantics/altIdentifier/doi/http://dx.doi.org/10.1158%2F0008-5472.CAN-12-2681  
dc.relation.alternativeid
info:eu-repo/semantics/altIdentifier/issn/0008-5472