Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells

Standard

Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells. / Knappe, Nathalie; Novak, Daniel; Weina, Kasia; Bernhardt, Mathias; Reith, Maike; Larribere, Lionel; Hölzel, Michael; Tüting, Thomas; Gebhardt, Christoffer; Umansky, Viktor; Utikal, Jochen.

in: Stem cells (Dayton, Ohio), Jahrgang 34, Nr. 4, 04.2016, S. 832-46.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Knappe, N, Novak, D, Weina, K, Bernhardt, M, Reith, M, Larribere, L, Hölzel, M, Tüting, T, Gebhardt, C, Umansky, V & Utikal, J 2016, 'Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells', Stem cells (Dayton, Ohio), Jg. 34, Nr. 4, S. 832-46. https://doi.org/10.1002/stem.2284

APA

Knappe, N., Novak, D., Weina, K., Bernhardt, M., Reith, M., Larribere, L., Hölzel, M., Tüting, T., Gebhardt, C., Umansky, V., & Utikal, J. (2016). Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells. Stem cells (Dayton, Ohio), 34(4), 832-46. https://doi.org/10.1002/stem.2284

Vancouver

Bibtex

@article{eac3af4f93d449b4a22920782b21a9ec,
title = "Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells",
abstract = "The combination of cancer-focused studies and research related to nuclear reprogramming has gained increasing importance since both processes-reprogramming towards pluripotency and malignant transformation-share essential features. Studies have revealed that incomplete reprogramming of somatic cells leads to malignant transformation indicating that epigenetic regulation associated with iPSC generation can drive cancer development [J Mol Cell Biol 2011;341-350; Cell 2012;151:1617-1632; Cell 2014;156:663-677]. However, so far it is unclear whether incomplete reprogramming also affects cancer cells and their function. In the context of melanoma, dedifferentiation correlates to therapy resistance in mouse studies and has been documented in melanoma patients [Nature 2012;490:412-416; Clin Cancer Res 2014;20:2498-2499]. Therefore, we sought to investigate directed dedifferentiation using incomplete reprogramming of melanoma cells. Using a murine model we investigated the effects of partial reprogramming on the cellular plasticity of melanoma cells. We demonstrate for the first time that induced partial reprogramming results in a reversible phenotype switch in melanoma cells. Partially reprogrammed cells at day 12 after transgene induction display elevated invasive potential in vitro and increased lung colonization in vivo. Additionally, using global gene expression analysis of partially reprogrammed cells, we identified SNAI3 as a novel invasion-related marker in human melanoma. SNAI3 expression correlates with tumor thickness in primary melanomas and thus, may be of prognostic value. In summary, we show that investigating intermediate states during the process of reprogramming melanoma cells can reveal novel insights into the pathogenesis of melanoma progression. We propose that deeper analysis of partially reprogrammed melanoma cells may contribute to identification of yet unknown signaling pathways that can drive melanoma progression.",
keywords = "Animals, Biomarkers, Tumor, Cell Dedifferentiation, Cellular Reprogramming, Gene Expression Regulation, Neoplastic, Humans, Induced Pluripotent Stem Cells, Melanoma, Mice, Mice, Transgenic, Neoplasm Invasiveness, Neoplasm Proteins, Neoplasms, Experimental, Signal Transduction, Snail Family Transcription Factors, Journal Article, Research Support, Non-U.S. Gov't",
author = "Nathalie Knappe and Daniel Novak and Kasia Weina and Mathias Bernhardt and Maike Reith and Lionel Larribere and Michael H{\"o}lzel and Thomas T{\"u}ting and Christoffer Gebhardt and Viktor Umansky and Jochen Utikal",
note = "{\textcopyright} 2016 AlphaMed Press.",
year = "2016",
month = apr,
doi = "10.1002/stem.2284",
language = "English",
volume = "34",
pages = "832--46",
journal = "STEM CELLS",
issn = "1066-5099",
publisher = "ALPHAMED PRESS",
number = "4",

}

RIS

TY - JOUR

T1 - Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells

AU - Knappe, Nathalie

AU - Novak, Daniel

AU - Weina, Kasia

AU - Bernhardt, Mathias

AU - Reith, Maike

AU - Larribere, Lionel

AU - Hölzel, Michael

AU - Tüting, Thomas

AU - Gebhardt, Christoffer

AU - Umansky, Viktor

AU - Utikal, Jochen

N1 - © 2016 AlphaMed Press.

PY - 2016/4

Y1 - 2016/4

N2 - The combination of cancer-focused studies and research related to nuclear reprogramming has gained increasing importance since both processes-reprogramming towards pluripotency and malignant transformation-share essential features. Studies have revealed that incomplete reprogramming of somatic cells leads to malignant transformation indicating that epigenetic regulation associated with iPSC generation can drive cancer development [J Mol Cell Biol 2011;341-350; Cell 2012;151:1617-1632; Cell 2014;156:663-677]. However, so far it is unclear whether incomplete reprogramming also affects cancer cells and their function. In the context of melanoma, dedifferentiation correlates to therapy resistance in mouse studies and has been documented in melanoma patients [Nature 2012;490:412-416; Clin Cancer Res 2014;20:2498-2499]. Therefore, we sought to investigate directed dedifferentiation using incomplete reprogramming of melanoma cells. Using a murine model we investigated the effects of partial reprogramming on the cellular plasticity of melanoma cells. We demonstrate for the first time that induced partial reprogramming results in a reversible phenotype switch in melanoma cells. Partially reprogrammed cells at day 12 after transgene induction display elevated invasive potential in vitro and increased lung colonization in vivo. Additionally, using global gene expression analysis of partially reprogrammed cells, we identified SNAI3 as a novel invasion-related marker in human melanoma. SNAI3 expression correlates with tumor thickness in primary melanomas and thus, may be of prognostic value. In summary, we show that investigating intermediate states during the process of reprogramming melanoma cells can reveal novel insights into the pathogenesis of melanoma progression. We propose that deeper analysis of partially reprogrammed melanoma cells may contribute to identification of yet unknown signaling pathways that can drive melanoma progression.

AB - The combination of cancer-focused studies and research related to nuclear reprogramming has gained increasing importance since both processes-reprogramming towards pluripotency and malignant transformation-share essential features. Studies have revealed that incomplete reprogramming of somatic cells leads to malignant transformation indicating that epigenetic regulation associated with iPSC generation can drive cancer development [J Mol Cell Biol 2011;341-350; Cell 2012;151:1617-1632; Cell 2014;156:663-677]. However, so far it is unclear whether incomplete reprogramming also affects cancer cells and their function. In the context of melanoma, dedifferentiation correlates to therapy resistance in mouse studies and has been documented in melanoma patients [Nature 2012;490:412-416; Clin Cancer Res 2014;20:2498-2499]. Therefore, we sought to investigate directed dedifferentiation using incomplete reprogramming of melanoma cells. Using a murine model we investigated the effects of partial reprogramming on the cellular plasticity of melanoma cells. We demonstrate for the first time that induced partial reprogramming results in a reversible phenotype switch in melanoma cells. Partially reprogrammed cells at day 12 after transgene induction display elevated invasive potential in vitro and increased lung colonization in vivo. Additionally, using global gene expression analysis of partially reprogrammed cells, we identified SNAI3 as a novel invasion-related marker in human melanoma. SNAI3 expression correlates with tumor thickness in primary melanomas and thus, may be of prognostic value. In summary, we show that investigating intermediate states during the process of reprogramming melanoma cells can reveal novel insights into the pathogenesis of melanoma progression. We propose that deeper analysis of partially reprogrammed melanoma cells may contribute to identification of yet unknown signaling pathways that can drive melanoma progression.

KW - Animals

KW - Biomarkers, Tumor

KW - Cell Dedifferentiation

KW - Cellular Reprogramming

KW - Gene Expression Regulation, Neoplastic

KW - Humans

KW - Induced Pluripotent Stem Cells

KW - Melanoma

KW - Mice

KW - Mice, Transgenic

KW - Neoplasm Invasiveness

KW - Neoplasm Proteins

KW - Neoplasms, Experimental

KW - Signal Transduction

KW - Snail Family Transcription Factors

KW - Journal Article

KW - Research Support, Non-U.S. Gov't

U2 - 10.1002/stem.2284

DO - 10.1002/stem.2284

M3 - SCORING: Journal article

C2 - 26753613

VL - 34

SP - 832

EP - 846

JO - STEM CELLS

JF - STEM CELLS

SN - 1066-5099

IS - 4

ER -