Cargando…

Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts

SIMPLE SUMMARY: We investigated the influence of autophagy-related variants in modulating colorectal cancer (CRC) risk through a meta-analysis of genome-wide association study (GWAS) data from four large European cohorts. We found that genetic variants within the DAPK2 and ATG5 loci were associated...

Descripción completa

Detalles Bibliográficos
Autores principales: Sainz, Juan, García-Verdejo, Francisco José, Martínez-Bueno, Manuel, Kumar, Abhishek, Sánchez-Maldonado, José Manuel, Díez-Villanueva, Anna, Vodičková, Ludmila, Vymetálková, Veronika, Martin Sánchez, Vicente, Da Silva Filho, Miguel Inacio, Sampaio-Marques, Belém, Brezina, Stefanie, Butterbach, Katja, ter Horst, Rob, Hoffmeister, Michael, Ludovico, Paula, Jurado, Manuel, Li, Yang, Sánchez-Rovira, Pedro, Netea, Mihai G., Gsur, Andrea, Vodička, Pavel, Moreno, Víctor, Hemminki, Kari, Brenner, Hermann, Chang-Claude, Jenny, Försti, Asta
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7998818/
https://www.ncbi.nlm.nih.gov/pubmed/33809172
http://dx.doi.org/10.3390/cancers13061258
_version_ 1783670639682387968
author Sainz, Juan
García-Verdejo, Francisco José
Martínez-Bueno, Manuel
Kumar, Abhishek
Sánchez-Maldonado, José Manuel
Díez-Villanueva, Anna
Vodičková, Ludmila
Vymetálková, Veronika
Martin Sánchez, Vicente
Da Silva Filho, Miguel Inacio
Sampaio-Marques, Belém
Brezina, Stefanie
Butterbach, Katja
ter Horst, Rob
Hoffmeister, Michael
Ludovico, Paula
Jurado, Manuel
Li, Yang
Sánchez-Rovira, Pedro
Netea, Mihai G.
Gsur, Andrea
Vodička, Pavel
Moreno, Víctor
Hemminki, Kari
Brenner, Hermann
Chang-Claude, Jenny
Försti, Asta
author_facet Sainz, Juan
García-Verdejo, Francisco José
Martínez-Bueno, Manuel
Kumar, Abhishek
Sánchez-Maldonado, José Manuel
Díez-Villanueva, Anna
Vodičková, Ludmila
Vymetálková, Veronika
Martin Sánchez, Vicente
Da Silva Filho, Miguel Inacio
Sampaio-Marques, Belém
Brezina, Stefanie
Butterbach, Katja
ter Horst, Rob
Hoffmeister, Michael
Ludovico, Paula
Jurado, Manuel
Li, Yang
Sánchez-Rovira, Pedro
Netea, Mihai G.
Gsur, Andrea
Vodička, Pavel
Moreno, Víctor
Hemminki, Kari
Brenner, Hermann
Chang-Claude, Jenny
Försti, Asta
author_sort Sainz, Juan
collection PubMed
description SIMPLE SUMMARY: We investigated the influence of autophagy-related variants in modulating colorectal cancer (CRC) risk through a meta-analysis of genome-wide association study (GWAS) data from four large European cohorts. We found that genetic variants within the DAPK2 and ATG5 loci were associated with CRC risk. This study also shed some light onto the functional mechanisms behind the observed associations and demonstrated the impact of DAPK2(rs11631973) and ATG5(rs546456) polymorphisms on the modulation of host immune responses, blood derived-cell counts and serum inflammatory protein levels, which might be involved in promoting cancer development. No effect of the DAPK2 and ATG5 polymorphisms on the autophagy flux was observed. ABSTRACT: The role of genetic variation in autophagy-related genes in modulating autophagy and cancer is poorly understood. Here, we comprehensively investigated the association of autophagy-related variants with colorectal cancer (CRC) risk and provide new insights about the molecular mechanisms underlying the associations. After meta-analysis of the genome-wide association study (GWAS) data from four independent European cohorts (8006 CRC cases and 7070 controls), two loci, DAPK2 (p = 2.19 × 10(−5)) and ATG5 (p = 6.28 × 10(−4)) were associated with the risk of CRC. Mechanistically, the DAPK2(rs11631973G) allele was associated with IL1 β levels after the stimulation of peripheral blood mononuclear cells (PBMCs) with Staphylococcus aureus (p = 0.002), CD24 + CD38 + CD27 + IgM + B cell levels in blood (p = 0.0038) and serum levels of en-RAGE (p = 0.0068). ATG5(rs546456T) allele was associated with TNF α and IL1 β levels after the stimulation of PBMCs with LPS (p = 0.0088 and p = 0.0076, respectively), CD14+CD16− cell levels in blood (p = 0.0068) and serum levels of CCL19 and cortisol (p = 0.0052 and p = 0.0074, respectively). Interestingly, no association with autophagy flux was observed. These results suggested an effect of the DAPK2 and ATG5 loci in the pathogenesis of CRC, likely through the modulation of host immune responses.
format Online
Article
Text
id pubmed-7998818
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-79988182021-03-28 Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts Sainz, Juan García-Verdejo, Francisco José Martínez-Bueno, Manuel Kumar, Abhishek Sánchez-Maldonado, José Manuel Díez-Villanueva, Anna Vodičková, Ludmila Vymetálková, Veronika Martin Sánchez, Vicente Da Silva Filho, Miguel Inacio Sampaio-Marques, Belém Brezina, Stefanie Butterbach, Katja ter Horst, Rob Hoffmeister, Michael Ludovico, Paula Jurado, Manuel Li, Yang Sánchez-Rovira, Pedro Netea, Mihai G. Gsur, Andrea Vodička, Pavel Moreno, Víctor Hemminki, Kari Brenner, Hermann Chang-Claude, Jenny Försti, Asta Cancers (Basel) Article SIMPLE SUMMARY: We investigated the influence of autophagy-related variants in modulating colorectal cancer (CRC) risk through a meta-analysis of genome-wide association study (GWAS) data from four large European cohorts. We found that genetic variants within the DAPK2 and ATG5 loci were associated with CRC risk. This study also shed some light onto the functional mechanisms behind the observed associations and demonstrated the impact of DAPK2(rs11631973) and ATG5(rs546456) polymorphisms on the modulation of host immune responses, blood derived-cell counts and serum inflammatory protein levels, which might be involved in promoting cancer development. No effect of the DAPK2 and ATG5 polymorphisms on the autophagy flux was observed. ABSTRACT: The role of genetic variation in autophagy-related genes in modulating autophagy and cancer is poorly understood. Here, we comprehensively investigated the association of autophagy-related variants with colorectal cancer (CRC) risk and provide new insights about the molecular mechanisms underlying the associations. After meta-analysis of the genome-wide association study (GWAS) data from four independent European cohorts (8006 CRC cases and 7070 controls), two loci, DAPK2 (p = 2.19 × 10(−5)) and ATG5 (p = 6.28 × 10(−4)) were associated with the risk of CRC. Mechanistically, the DAPK2(rs11631973G) allele was associated with IL1 β levels after the stimulation of peripheral blood mononuclear cells (PBMCs) with Staphylococcus aureus (p = 0.002), CD24 + CD38 + CD27 + IgM + B cell levels in blood (p = 0.0038) and serum levels of en-RAGE (p = 0.0068). ATG5(rs546456T) allele was associated with TNF α and IL1 β levels after the stimulation of PBMCs with LPS (p = 0.0088 and p = 0.0076, respectively), CD14+CD16− cell levels in blood (p = 0.0068) and serum levels of CCL19 and cortisol (p = 0.0052 and p = 0.0074, respectively). Interestingly, no association with autophagy flux was observed. These results suggested an effect of the DAPK2 and ATG5 loci in the pathogenesis of CRC, likely through the modulation of host immune responses. MDPI 2021-03-12 /pmc/articles/PMC7998818/ /pubmed/33809172 http://dx.doi.org/10.3390/cancers13061258 Text en © 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Sainz, Juan
García-Verdejo, Francisco José
Martínez-Bueno, Manuel
Kumar, Abhishek
Sánchez-Maldonado, José Manuel
Díez-Villanueva, Anna
Vodičková, Ludmila
Vymetálková, Veronika
Martin Sánchez, Vicente
Da Silva Filho, Miguel Inacio
Sampaio-Marques, Belém
Brezina, Stefanie
Butterbach, Katja
ter Horst, Rob
Hoffmeister, Michael
Ludovico, Paula
Jurado, Manuel
Li, Yang
Sánchez-Rovira, Pedro
Netea, Mihai G.
Gsur, Andrea
Vodička, Pavel
Moreno, Víctor
Hemminki, Kari
Brenner, Hermann
Chang-Claude, Jenny
Försti, Asta
Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title_full Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title_fullStr Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title_full_unstemmed Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title_short Polymorphisms within Autophagy-Related Genes Influence the Risk of Developing Colorectal Cancer: A Meta-Analysis of Four Large Cohorts
title_sort polymorphisms within autophagy-related genes influence the risk of developing colorectal cancer: a meta-analysis of four large cohorts
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7998818/
https://www.ncbi.nlm.nih.gov/pubmed/33809172
http://dx.doi.org/10.3390/cancers13061258
work_keys_str_mv AT sainzjuan polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT garciaverdejofranciscojose polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT martinezbuenomanuel polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT kumarabhishek polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT sanchezmaldonadojosemanuel polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT diezvillanuevaanna polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT vodickovaludmila polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT vymetalkovaveronika polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT martinsanchezvicente polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT dasilvafilhomiguelinacio polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT sampaiomarquesbelem polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT brezinastefanie polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT butterbachkatja polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT terhorstrob polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT hoffmeistermichael polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT ludovicopaula polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT juradomanuel polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT liyang polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT sanchezrovirapedro polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT neteamihaig polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT gsurandrea polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT vodickapavel polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT morenovictor polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT hemminkikari polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT brennerhermann polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT changclaudejenny polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts
AT forstiasta polymorphismswithinautophagyrelatedgenesinfluencetheriskofdevelopingcolorectalcancerametaanalysisoffourlargecohorts