Cargando…

Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells

The pivotal role of cancer initiating stem cells (CSCs) in tumor initiation, growth, metastasis and drug resistance has led to the postulation of a ‘total cancer therapy’ paradigm, which involves targeting both cancer cells and CSCs for effective therapy. However, the progress in identifying drugs f...

Descripción completa

Detalles Bibliográficos
Autores principales: Quarni, Waise, Dutta, Rinku, Green, Ryan, Katiri, Sandhyabanu, Patel, Bhaumik, Mohapatra, Shyam S., Mohapatra, Subhra
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6811578/
https://www.ncbi.nlm.nih.gov/pubmed/31645574
http://dx.doi.org/10.1038/s41598-019-50917-3
_version_ 1783462493348167680
author Quarni, Waise
Dutta, Rinku
Green, Ryan
Katiri, Sandhyabanu
Patel, Bhaumik
Mohapatra, Shyam S.
Mohapatra, Subhra
author_facet Quarni, Waise
Dutta, Rinku
Green, Ryan
Katiri, Sandhyabanu
Patel, Bhaumik
Mohapatra, Shyam S.
Mohapatra, Subhra
author_sort Quarni, Waise
collection PubMed
description The pivotal role of cancer initiating stem cells (CSCs) in tumor initiation, growth, metastasis and drug resistance has led to the postulation of a ‘total cancer therapy’ paradigm, which involves targeting both cancer cells and CSCs for effective therapy. However, the progress in identifying drugs for total cancer therapy has been limited. Herein, we show for the first time that mithramycin A (Mit-A) can successfully inhibit CSC proliferation, in addition to inhibiting bulk cancer cells in a model of colorectal cancer (CRC), the second leading cause of death among men and women in the United States. To this end, a polymeric nanofiber scaffold culture system was established to develop 3D tumor organoids (tumoroids) from CRC cell lines such as HT29, HCT116, KM12, CT26 and MC38 as well as ex vivo mouse tumors. These tumoroids possessed increased expression of CSC markers and transcription factors, expanded the number of CSCs in culture and increased CSC functional properties measured by aldehyde dehydrogenase activity. Screening of an NCI library of FDA approved drugs led to the identification of Mit-A as a potential total cancer therapy drug. In both sphere and tumoroid culture, Mit-A inhibits cancer growth by reducing the expression of cancer stemness markers. In addition, Mit-A inhibits the expression of SP1, a previously known target in CRCs. Moreover, Mit-A significantly reduces growth of tumoroids in ex vivo cultures and CRC tumor growth in vivo. Finally, a dose-dependent treatment on CRC cells indicate that Mit-A significantly induces the cell death and PARP-cleavage of both CSC and non-CSC cells. Taken together the results of these in vitro, ex vivo and in vivo studies lead to the inference that Mit-A is a promising drug candidate for total cancer therapy of CRCs.
format Online
Article
Text
id pubmed-6811578
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-68115782019-10-25 Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells Quarni, Waise Dutta, Rinku Green, Ryan Katiri, Sandhyabanu Patel, Bhaumik Mohapatra, Shyam S. Mohapatra, Subhra Sci Rep Article The pivotal role of cancer initiating stem cells (CSCs) in tumor initiation, growth, metastasis and drug resistance has led to the postulation of a ‘total cancer therapy’ paradigm, which involves targeting both cancer cells and CSCs for effective therapy. However, the progress in identifying drugs for total cancer therapy has been limited. Herein, we show for the first time that mithramycin A (Mit-A) can successfully inhibit CSC proliferation, in addition to inhibiting bulk cancer cells in a model of colorectal cancer (CRC), the second leading cause of death among men and women in the United States. To this end, a polymeric nanofiber scaffold culture system was established to develop 3D tumor organoids (tumoroids) from CRC cell lines such as HT29, HCT116, KM12, CT26 and MC38 as well as ex vivo mouse tumors. These tumoroids possessed increased expression of CSC markers and transcription factors, expanded the number of CSCs in culture and increased CSC functional properties measured by aldehyde dehydrogenase activity. Screening of an NCI library of FDA approved drugs led to the identification of Mit-A as a potential total cancer therapy drug. In both sphere and tumoroid culture, Mit-A inhibits cancer growth by reducing the expression of cancer stemness markers. In addition, Mit-A inhibits the expression of SP1, a previously known target in CRCs. Moreover, Mit-A significantly reduces growth of tumoroids in ex vivo cultures and CRC tumor growth in vivo. Finally, a dose-dependent treatment on CRC cells indicate that Mit-A significantly induces the cell death and PARP-cleavage of both CSC and non-CSC cells. Taken together the results of these in vitro, ex vivo and in vivo studies lead to the inference that Mit-A is a promising drug candidate for total cancer therapy of CRCs. Nature Publishing Group UK 2019-10-23 /pmc/articles/PMC6811578/ /pubmed/31645574 http://dx.doi.org/10.1038/s41598-019-50917-3 Text en © The Author(s) 2019 Open Access This 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 license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license 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 license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Quarni, Waise
Dutta, Rinku
Green, Ryan
Katiri, Sandhyabanu
Patel, Bhaumik
Mohapatra, Shyam S.
Mohapatra, Subhra
Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title_full Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title_fullStr Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title_full_unstemmed Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title_short Mithramycin A Inhibits Colorectal Cancer Growth by Targeting Cancer Stem Cells
title_sort mithramycin a inhibits colorectal cancer growth by targeting cancer stem cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6811578/
https://www.ncbi.nlm.nih.gov/pubmed/31645574
http://dx.doi.org/10.1038/s41598-019-50917-3
work_keys_str_mv AT quarniwaise mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT duttarinku mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT greenryan mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT katirisandhyabanu mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT patelbhaumik mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT mohapatrashyams mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells
AT mohapatrasubhra mithramycinainhibitscolorectalcancergrowthbytargetingcancerstemcells