Cargando…

Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival

BACKGROUND: Progression of colorectal cancer (CRC), a leading cause of cancer-related death worldwide, is driven by colorectal cancer stem cells (CR-CSCs), which are regulated by endogenous and microenvironmental signals. Interleukin (IL)-30 has proven to be crucial for CSC viability and tumor progr...

Descripción completa

Detalles Bibliográficos
Autores principales: D'Antonio, Luigi, Fieni, Cristiano, Ciummo, Stefania Livia, Vespa, Simone, Lotti, Lavinia, Sorrentino, Carlo, Di Carlo, Emma
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10030651/
https://www.ncbi.nlm.nih.gov/pubmed/36927528
http://dx.doi.org/10.1136/jitc-2022-006056
_version_ 1784910425835438080
author D'Antonio, Luigi
Fieni, Cristiano
Ciummo, Stefania Livia
Vespa, Simone
Lotti, Lavinia
Sorrentino, Carlo
Di Carlo, Emma
author_facet D'Antonio, Luigi
Fieni, Cristiano
Ciummo, Stefania Livia
Vespa, Simone
Lotti, Lavinia
Sorrentino, Carlo
Di Carlo, Emma
author_sort D'Antonio, Luigi
collection PubMed
description BACKGROUND: Progression of colorectal cancer (CRC), a leading cause of cancer-related death worldwide, is driven by colorectal cancer stem cells (CR-CSCs), which are regulated by endogenous and microenvironmental signals. Interleukin (IL)-30 has proven to be crucial for CSC viability and tumor progression. Whether it is involved in CRC tumorigenesis and impacts clinical behavior is unknown. METHODS: IL30 production and functions, in stem and non-stem CRC cells, were determined by western blot, immunoelectron microscopy, flow cytometry, cell viability and sphere formation assays. CRISPR/Cas9-mediated deletion of the IL30 gene, RNA-Seq and implantation of IL30 gene transfected or deleted CR-CSCs in NSG mice allowed to investigate IL30’s role in CRC oncogenesis. Bioinformatics and immunopathology of CRC samples highlighted the clinical implications. RESULTS: We demonstrated that both CR-CSCs and CRC cells express membrane-anchored IL30 that regulates their self-renewal, via WNT5A and RAB33A, and/or proliferation and migration, primarily by upregulating CXCR4 via STAT3, which are suppressed by IL30 gene deletion, along with WNT and RAS pathways. Deletion of IL30 gene downregulates the expression of proteases, such as MMP2 and MMP13, chemokine receptors, mostly CCR7, CCR3 and CXCR4, and growth and inflammatory mediators, including ANGPT2, CXCL10, EPO, IGF1 and EGF. These factors contribute to IL30-driven CR-CSC and CRC cell expansion, which is abrogated by their selective blockade. IL30 gene deleted CR-CSCs displayed reduced tumorigenicity and gave rise to slow-growing and low metastatic tumors in 80% of mice, which survived much longer than controls. Bioinformatics and CIBERSORTx of the ‘Colorectal Adenocarcinoma TCGA Nature 2012’ collection, and morphometric assessment of IL30 expression in clinical CRC samples revealed that the lack of IL30 in CRC and infiltrating leucocytes correlates with prolonged overall survival. CONCLUSIONS: IL30 is a new CRC driver, since its inactivation, which disables oncogenic pathways and multiple autocrine loops, inhibits CR-CSC tumorigenicity and metastatic ability. The development of CRISPR/Cas9-mediated targeting of IL30 could improve the current therapeutic landscape of CRC.
format Online
Article
Text
id pubmed-10030651
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-100306512023-03-23 Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival D'Antonio, Luigi Fieni, Cristiano Ciummo, Stefania Livia Vespa, Simone Lotti, Lavinia Sorrentino, Carlo Di Carlo, Emma J Immunother Cancer Basic Tumor Immunology BACKGROUND: Progression of colorectal cancer (CRC), a leading cause of cancer-related death worldwide, is driven by colorectal cancer stem cells (CR-CSCs), which are regulated by endogenous and microenvironmental signals. Interleukin (IL)-30 has proven to be crucial for CSC viability and tumor progression. Whether it is involved in CRC tumorigenesis and impacts clinical behavior is unknown. METHODS: IL30 production and functions, in stem and non-stem CRC cells, were determined by western blot, immunoelectron microscopy, flow cytometry, cell viability and sphere formation assays. CRISPR/Cas9-mediated deletion of the IL30 gene, RNA-Seq and implantation of IL30 gene transfected or deleted CR-CSCs in NSG mice allowed to investigate IL30’s role in CRC oncogenesis. Bioinformatics and immunopathology of CRC samples highlighted the clinical implications. RESULTS: We demonstrated that both CR-CSCs and CRC cells express membrane-anchored IL30 that regulates their self-renewal, via WNT5A and RAB33A, and/or proliferation and migration, primarily by upregulating CXCR4 via STAT3, which are suppressed by IL30 gene deletion, along with WNT and RAS pathways. Deletion of IL30 gene downregulates the expression of proteases, such as MMP2 and MMP13, chemokine receptors, mostly CCR7, CCR3 and CXCR4, and growth and inflammatory mediators, including ANGPT2, CXCL10, EPO, IGF1 and EGF. These factors contribute to IL30-driven CR-CSC and CRC cell expansion, which is abrogated by their selective blockade. IL30 gene deleted CR-CSCs displayed reduced tumorigenicity and gave rise to slow-growing and low metastatic tumors in 80% of mice, which survived much longer than controls. Bioinformatics and CIBERSORTx of the ‘Colorectal Adenocarcinoma TCGA Nature 2012’ collection, and morphometric assessment of IL30 expression in clinical CRC samples revealed that the lack of IL30 in CRC and infiltrating leucocytes correlates with prolonged overall survival. CONCLUSIONS: IL30 is a new CRC driver, since its inactivation, which disables oncogenic pathways and multiple autocrine loops, inhibits CR-CSC tumorigenicity and metastatic ability. The development of CRISPR/Cas9-mediated targeting of IL30 could improve the current therapeutic landscape of CRC. BMJ Publishing Group 2023-03-16 /pmc/articles/PMC10030651/ /pubmed/36927528 http://dx.doi.org/10.1136/jitc-2022-006056 Text en © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Basic Tumor Immunology
D'Antonio, Luigi
Fieni, Cristiano
Ciummo, Stefania Livia
Vespa, Simone
Lotti, Lavinia
Sorrentino, Carlo
Di Carlo, Emma
Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title_full Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title_fullStr Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title_full_unstemmed Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title_short Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival
title_sort inactivation of interleukin-30 in colon cancer stem cells via crispr/cas9 genome editing inhibits their oncogenicity and improves host survival
topic Basic Tumor Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10030651/
https://www.ncbi.nlm.nih.gov/pubmed/36927528
http://dx.doi.org/10.1136/jitc-2022-006056
work_keys_str_mv AT dantonioluigi inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT fienicristiano inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT ciummostefanialivia inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT vespasimone inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT lottilavinia inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT sorrentinocarlo inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival
AT dicarloemma inactivationofinterleukin30incoloncancerstemcellsviacrisprcas9genomeeditinginhibitstheironcogenicityandimproveshostsurvival