Cargando…

Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence

BACKGROUND: The oncogene LSF (encoded by TFCP2) has been proposed as a novel therapeutic target for multiple cancers. LSF overexpression in patient tumors correlates with poor prognosis in particular for both hepatocellular carcinoma and colorectal cancer. The limited treatment outcomes for these di...

Descripción completa

Detalles Bibliográficos
Autores principales: Willoughby, Jennifer L. S., George, Kelly, Roberto, Mark P., Chin, Hang Gyeong, Stoiber, Patrick, Shin, Hyunjin, Pedamallu, Chandra Sekhar, Schaus, Scott E., Fitzgerald, Kevin, Shah, Jagesh, Hansen, Ulla
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7296649/
https://www.ncbi.nlm.nih.gov/pubmed/32539694
http://dx.doi.org/10.1186/s12885-020-07039-1
_version_ 1783546874065584128
author Willoughby, Jennifer L. S.
George, Kelly
Roberto, Mark P.
Chin, Hang Gyeong
Stoiber, Patrick
Shin, Hyunjin
Pedamallu, Chandra Sekhar
Schaus, Scott E.
Fitzgerald, Kevin
Shah, Jagesh
Hansen, Ulla
author_facet Willoughby, Jennifer L. S.
George, Kelly
Roberto, Mark P.
Chin, Hang Gyeong
Stoiber, Patrick
Shin, Hyunjin
Pedamallu, Chandra Sekhar
Schaus, Scott E.
Fitzgerald, Kevin
Shah, Jagesh
Hansen, Ulla
author_sort Willoughby, Jennifer L. S.
collection PubMed
description BACKGROUND: The oncogene LSF (encoded by TFCP2) has been proposed as a novel therapeutic target for multiple cancers. LSF overexpression in patient tumors correlates with poor prognosis in particular for both hepatocellular carcinoma and colorectal cancer. The limited treatment outcomes for these diseases and disappointing clinical results, in particular, for hepatocellular carcinoma in molecularly targeted therapies targeting cellular receptors and kinases, underscore the need for molecularly targeting novel mechanisms. LSF small molecule inhibitors, Factor Quinolinone Inhibitors (FQIs), have exhibited robust anti-tumor activity in multiple pre-clinical models, with no observable toxicity. METHODS: To understand how the LSF inhibitors impact cancer cell proliferation, we characterized the cellular phenotypes that result from loss of LSF activity. Cell proliferation and cell cycle progression were analyzed, using HeLa cells as a model cancer cell line responsive to FQI1. Cell cycle progression was studied either by time lapse microscopy or by bulk synchronization of cell populations to ensure accuracy in interpretation of the outcomes. In order to test for biological specificity of targeting LSF by FQI1, results were compared after treatment with either FQI1 or siRNA targeting LSF. RESULTS: Highly similar cellular phenotypes are observed upon treatments with FQI1 and siRNA targeting LSF. Along with similar effects on two cellular biomarkers, inhibition of LSF activity by either mechanism induced a strong delay or arrest prior to metaphase as cells progressed through mitosis, with condensed, but unaligned, chromosomes. This mitotic disruption in both cases resulted in improper cellular division leading to multiple outcomes: multi-nucleation, apoptosis, and cellular senescence. CONCLUSIONS: These data strongly support that cellular phenotypes observed upon FQI1 treatment are due specifically to the loss of LSF activity. Specific inhibition of LSF by either small molecules or siRNA results in severe mitotic defects, leading to cell death or senescence - consequences that are desirable in combating cancer. Taken together, these findings confirm that LSF is a promising target for cancer treatment. Furthermore, this study provides further support for developing FQIs or other LSF inhibitory strategies as treatment for LSF-related cancers with high unmet medical needs.
format Online
Article
Text
id pubmed-7296649
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-72966492020-06-16 Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence Willoughby, Jennifer L. S. George, Kelly Roberto, Mark P. Chin, Hang Gyeong Stoiber, Patrick Shin, Hyunjin Pedamallu, Chandra Sekhar Schaus, Scott E. Fitzgerald, Kevin Shah, Jagesh Hansen, Ulla BMC Cancer Research Article BACKGROUND: The oncogene LSF (encoded by TFCP2) has been proposed as a novel therapeutic target for multiple cancers. LSF overexpression in patient tumors correlates with poor prognosis in particular for both hepatocellular carcinoma and colorectal cancer. The limited treatment outcomes for these diseases and disappointing clinical results, in particular, for hepatocellular carcinoma in molecularly targeted therapies targeting cellular receptors and kinases, underscore the need for molecularly targeting novel mechanisms. LSF small molecule inhibitors, Factor Quinolinone Inhibitors (FQIs), have exhibited robust anti-tumor activity in multiple pre-clinical models, with no observable toxicity. METHODS: To understand how the LSF inhibitors impact cancer cell proliferation, we characterized the cellular phenotypes that result from loss of LSF activity. Cell proliferation and cell cycle progression were analyzed, using HeLa cells as a model cancer cell line responsive to FQI1. Cell cycle progression was studied either by time lapse microscopy or by bulk synchronization of cell populations to ensure accuracy in interpretation of the outcomes. In order to test for biological specificity of targeting LSF by FQI1, results were compared after treatment with either FQI1 or siRNA targeting LSF. RESULTS: Highly similar cellular phenotypes are observed upon treatments with FQI1 and siRNA targeting LSF. Along with similar effects on two cellular biomarkers, inhibition of LSF activity by either mechanism induced a strong delay or arrest prior to metaphase as cells progressed through mitosis, with condensed, but unaligned, chromosomes. This mitotic disruption in both cases resulted in improper cellular division leading to multiple outcomes: multi-nucleation, apoptosis, and cellular senescence. CONCLUSIONS: These data strongly support that cellular phenotypes observed upon FQI1 treatment are due specifically to the loss of LSF activity. Specific inhibition of LSF by either small molecules or siRNA results in severe mitotic defects, leading to cell death or senescence - consequences that are desirable in combating cancer. Taken together, these findings confirm that LSF is a promising target for cancer treatment. Furthermore, this study provides further support for developing FQIs or other LSF inhibitory strategies as treatment for LSF-related cancers with high unmet medical needs. BioMed Central 2020-06-15 /pmc/articles/PMC7296649/ /pubmed/32539694 http://dx.doi.org/10.1186/s12885-020-07039-1 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research Article
Willoughby, Jennifer L. S.
George, Kelly
Roberto, Mark P.
Chin, Hang Gyeong
Stoiber, Patrick
Shin, Hyunjin
Pedamallu, Chandra Sekhar
Schaus, Scott E.
Fitzgerald, Kevin
Shah, Jagesh
Hansen, Ulla
Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title_full Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title_fullStr Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title_full_unstemmed Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title_short Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
title_sort targeting the oncogene lsf with either the small molecule inhibitor fqi1 or sirna causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7296649/
https://www.ncbi.nlm.nih.gov/pubmed/32539694
http://dx.doi.org/10.1186/s12885-020-07039-1
work_keys_str_mv AT willoughbyjenniferls targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT georgekelly targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT robertomarkp targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT chinhanggyeong targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT stoiberpatrick targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT shinhyunjin targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT pedamalluchandrasekhar targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT schausscotte targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT fitzgeraldkevin targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT shahjagesh targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence
AT hansenulla targetingtheoncogenelsfwitheitherthesmallmoleculeinhibitorfqi1orsirnacausesmitoticdelayswithunalignedchromosomesresultingincelldeathorsenescence