[HTML][HTML] Notch3 signaling-mediated melanoma–endothelial crosstalk regulates melanoma stem-like cell homeostasis and niche morphogenesis

MY Hsu, MH Yang, CI Schnegg, S Hwang… - Laboratory …, 2017 - nature.com
MY Hsu, MH Yang, CI Schnegg, S Hwang, B Ryu, RM Alani
Laboratory investigation, 2017nature.com
Melanoma is among the most virulent cancers, owing to its propensity to metastasize and its
resistance to current therapies. The treatment failure is largely attributed to tumor
heterogeneity, particularly subpopulations possessing stem cell-like properties, ie,
melanoma stem-like cells (MSLCs). Evidence indicates that the MSLC phenotype is
malleable and may be acquired by non-MSLCs through phenotypic switching upon
appropriate stimuli, the so–called 'dynamic stemness'. Since the phenotypic characteristics …
Abstract
Melanoma is among the most virulent cancers, owing to its propensity to metastasize and its resistance to current therapies. The treatment failure is largely attributed to tumor heterogeneity, particularly subpopulations possessing stem cell-like properties, ie, melanoma stem-like cells (MSLCs). Evidence indicates that the MSLC phenotype is malleable and may be acquired by non-MSLCs through phenotypic switching upon appropriate stimuli, the so–called ‘dynamic stemness’. Since the phenotypic characteristics and functional integrity of MSLCs depend on their vascular niche, using a two-dimensional (2D) melanoma–endothelium co-culture model, where the MSLC niche is recapitulated in vitro, we identified Notch3 signaling pathway as a micro-environmental cue governing MSLC phenotypic plasticity via pathway-specific gene expression arrays. Accordingly, lentiviral shRNA-mediated Notch3 knockdown (KD) in melanoma cell lines exhibiting high levels of endogenous Notch3 led to retarded/abolished tumorigenicity in vivo through both depleting MSLC fractions, evinced by MSLC marker downregulation (eg, CD133 and CD271); and impeding the MSLC niche, corroborated by the attenuated tumor angiogenesis as well as vasculogenic mimicry. In contrast, Notch3 KD affected neither tumor growth nor MSLC subsets in a melanoma cell line with relatively low endogenous Notch3 expression. Thus, Notch3 signaling may facilitate MSLC plasticity and niche morphogenesis in a cell context-dependent manner. Our findings illustrate Notch3 as a molecular switch driving melanoma heterogeneity, and provide the biological rationale for Notch inhibition as a promising therapeutic option.
nature.com