Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cells ; 29(11): 1752-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21948558

ABSTRACT

Mouse and human somatic cells can either be reprogrammed to a pluripotent state or converted to another lineage with a combination of transcription factors suggesting that lineage commitment is a reversible process. Here we show that only one factor, the active intracellular form of Notch1, is sufficient to convert mature pigmented epidermal-derived melanocytes into functional multipotent neural crest (NC) stem-like cells. These induced NC stem cells (iNCSCs) proliferate as spheres under stem cell media conditions, re-express NC-related genes, and differentiate into multiple NC-derived mesenchymal and neuronal lineages. Moreover, iNCSCs are highly migratory and functional in vivo. These results demonstrate that mature melanocytes can be reprogrammed toward their primitive NC cell precursors through the activation of a single stem cell-related pathway. Reprogramming of melanocytes to iNCSCs may provide an alternate source of NCSCs for neuroregenerative applications.


Subject(s)
Cellular Reprogramming/physiology , Melanocytes/cytology , Melanocytes/metabolism , Neural Crest/cytology , Neural Stem Cells/cytology , Receptor, Notch1/metabolism , Stem Cells/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Cellular Reprogramming/genetics , Chick Embryo , Humans , Neural Stem Cells/metabolism , Receptor, Notch1/genetics , Stem Cells/metabolism
2.
Am J Pathol ; 174(6): 2367-77, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19389934

ABSTRACT

Melanocytic nevi frequently harbor oncogenic BRAF mutations, but only a minority progress to melanoma. In human melanocytes, persistent BRAF(V600E) expression triggers oncogene-induced senescence, which implies that bypass of oncogene-induced senescence is necessary for malignant transformation of melanocytes. We show that a subpopulation of primary human melanocytes with persistent expression of BRAF(V600E) do not enter oncogene-induced senescence, but instead survive despite heightened MAPK activity. Disruption of the p53 pathway using short-hairpin RNA initiated rapid growth of these V600E(+) melanocytes in vitro. The resultant V600E(+)/p53(sh) melanocytes grew anchorage-independently in soft agar, formed pigmented lesions reminiscent of in situ melanoma in artificial skin reconstructs, and were weakly tumorigenic in vivo. Array comparative genomic hybridization analysis demonstrated that the transformed melanocytes acquired a substantial deletion in chromosome 13, which encodes the Rb1 tumor suppressor gene. Gene expression profiling study of nevi and melanomas showed that p53 target genes were differentially expressed in melanomas compared with nevi, suggesting a dysfunctional p53 pathway in melanoma in vivo. In summary, these data demonstrate that a subpopulation of melanocytes possesses the ability to survive BRAF(V600E)-induced senescence, and suggest that p53 inactivation may promote malignant transformation of these cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Melanocytes/pathology , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cells, Cultured , Cellular Senescence/genetics , Comparative Genomic Hybridization , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, SCID , Mutation , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Retinoblastoma Protein/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...