TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2

The transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overex...

Full description

Bibliographic Details
Main Authors: Amy T Ku, Timothy M Shaver, Ajay S Rao, Jeffrey M Howard, Christine N Rodriguez, Qi Miao, Gloria Garcia, Diep Le, Diane Yang, Malgorzata Borowiak, Daniel N Cohen, Vida Chitsazzadeh, Abdul H Diwan, Kenneth Y Tsai, Hoang Nguyen
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2017-05-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/23242
_version_ 1811181118494343168
author Amy T Ku
Timothy M Shaver
Ajay S Rao
Jeffrey M Howard
Christine N Rodriguez
Qi Miao
Gloria Garcia
Diep Le
Diane Yang
Malgorzata Borowiak
Daniel N Cohen
Vida Chitsazzadeh
Abdul H Diwan
Kenneth Y Tsai
Hoang Nguyen
author_facet Amy T Ku
Timothy M Shaver
Ajay S Rao
Jeffrey M Howard
Christine N Rodriguez
Qi Miao
Gloria Garcia
Diep Le
Diane Yang
Malgorzata Borowiak
Daniel N Cohen
Vida Chitsazzadeh
Abdul H Diwan
Kenneth Y Tsai
Hoang Nguyen
author_sort Amy T Ku
collection DOAJ
description The transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overexpression increases tumor incidence, tumor multiplicity, and malignant progression in the chemically induced mouse model of skin SCC. Additionally, we show that downregulation of TCF7L1 and its paralogue TCF7L2 reduces tumor growth in a xenograft model of human skin SCC. Using separation-of-function mutants, we show that TCF7L1 promotes tumor growth, enhances cell migration, and overrides oncogenic RAS-induced senescence independently of its interaction with β-catenin. Through transcriptome profiling and combined gain- and loss-of-function studies, we identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. Our findings establish a tumor-promoting role for TCF7L1 in skin and elucidate the mechanisms underlying its tumorigenic capacity.
first_indexed 2024-04-11T09:13:03Z
format Article
id doaj.art-b98fd3e38ef8466c89853859555a1e85
institution Directory Open Access Journal
issn 2050-084X
language English
last_indexed 2024-04-11T09:13:03Z
publishDate 2017-05-01
publisher eLife Sciences Publications Ltd
record_format Article
series eLife
spelling doaj.art-b98fd3e38ef8466c89853859555a1e852022-12-22T04:32:26ZengeLife Sciences Publications LtdeLife2050-084X2017-05-01610.7554/eLife.23242TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2Amy T Ku0Timothy M Shaver1Ajay S Rao2Jeffrey M Howard3Christine N Rodriguez4Qi Miao5Gloria Garcia6Diep Le7Diane Yang8Malgorzata Borowiak9Daniel N Cohen10Vida Chitsazzadeh11Abdul H Diwan12Kenneth Y Tsai13Hoang Nguyen14https://orcid.org/0000-0002-1091-7483Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United States; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United States; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United States; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United States; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States; Program in Developmental Biology, Baylor College of Medicine, Houston, United States; McNair Medical Institute, Baylor College of Medicine, Houston, United StatesDepartment of Pathology and Immunology, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, United StatesDepartment of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, United StatesDepartment of Dermatology, Baylor College of Medicine, Houston, United StatesDepartment of Tumor Biology, Moffitt Cancer Center, Tampa, United States; Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, United StatesStem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, United States; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, United States; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States; Program in Developmental Biology, Baylor College of Medicine, Houston, United States; Department of Dermatology, Baylor College of Medicine, Houston, United States; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, United StatesThe transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overexpression increases tumor incidence, tumor multiplicity, and malignant progression in the chemically induced mouse model of skin SCC. Additionally, we show that downregulation of TCF7L1 and its paralogue TCF7L2 reduces tumor growth in a xenograft model of human skin SCC. Using separation-of-function mutants, we show that TCF7L1 promotes tumor growth, enhances cell migration, and overrides oncogenic RAS-induced senescence independently of its interaction with β-catenin. Through transcriptome profiling and combined gain- and loss-of-function studies, we identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. Our findings establish a tumor-promoting role for TCF7L1 in skin and elucidate the mechanisms underlying its tumorigenic capacity.https://elifesciences.org/articles/23242TCF7L1skin squamous cell carcinomacell migrationoncogene-induced senescence
spellingShingle Amy T Ku
Timothy M Shaver
Ajay S Rao
Jeffrey M Howard
Christine N Rodriguez
Qi Miao
Gloria Garcia
Diep Le
Diane Yang
Malgorzata Borowiak
Daniel N Cohen
Vida Chitsazzadeh
Abdul H Diwan
Kenneth Y Tsai
Hoang Nguyen
TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
eLife
TCF7L1
skin squamous cell carcinoma
cell migration
oncogene-induced senescence
title TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
title_full TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
title_fullStr TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
title_full_unstemmed TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
title_short TCF7L1 promotes skin tumorigenesis independently of β-catenin through induction of LCN2
title_sort tcf7l1 promotes skin tumorigenesis independently of β catenin through induction of lcn2
topic TCF7L1
skin squamous cell carcinoma
cell migration
oncogene-induced senescence
url https://elifesciences.org/articles/23242
work_keys_str_mv AT amytku tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT timothymshaver tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT ajaysrao tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT jeffreymhoward tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT christinenrodriguez tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT qimiao tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT gloriagarcia tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT dieple tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT dianeyang tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT malgorzataborowiak tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT danielncohen tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT vidachitsazzadeh tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT abdulhdiwan tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT kennethytsai tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2
AT hoangnguyen tcf7l1promotesskintumorigenesisindependentlyofbcateninthroughinductionoflcn2