Cargando…

Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing

BACKGROUND: Acute lymphoblastic leukemia (ALL), the most common childhood malignancy, is characterized by recurring structural chromosomal alterations and genetic alterations, whose detection is critical in diagnosis, risk stratification and prognostication. However, the genetic mechanisms that give...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Honghong, Wang, Hongsheng, Qian, Xiaowen, Gao, Shuai, Xia, Jieqi, Liu, Junwen, Cheng, Yanqin, Man, Jie, Zhai, Xiaowen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7068927/
https://www.ncbi.nlm.nih.gov/pubmed/32164600
http://dx.doi.org/10.1186/s12885-020-6709-7
_version_ 1783505673373351936
author Zhang, Honghong
Wang, Hongsheng
Qian, Xiaowen
Gao, Shuai
Xia, Jieqi
Liu, Junwen
Cheng, Yanqin
Man, Jie
Zhai, Xiaowen
author_facet Zhang, Honghong
Wang, Hongsheng
Qian, Xiaowen
Gao, Shuai
Xia, Jieqi
Liu, Junwen
Cheng, Yanqin
Man, Jie
Zhai, Xiaowen
author_sort Zhang, Honghong
collection PubMed
description BACKGROUND: Acute lymphoblastic leukemia (ALL), the most common childhood malignancy, is characterized by recurring structural chromosomal alterations and genetic alterations, whose detection is critical in diagnosis, risk stratification and prognostication. However, the genetic mechanisms that give rise to ALL remain poorly understood. METHODS: Using next-generation sequencing (NGS) in matched germline and tumor samples from 140 pediatric Chinese patients with ALL, we landscaped the gene mutations and estimated the mutation frequencies in this disease. RESULTS: Our results showed that the top driver oncogenes having a mutation prevalence over 5% in childhood ALL included KRAS (8.76%), NRAS (6.4%), FLT3 (5.7%) and KMT2D (5.0%). While the most frequently mutated genes were KRAS, NRAS and FLT3 in B cell ALL (B-ALL), the most common mutations were enriched in NOTCH1 (23.1%), FBXW7 (23.1%) and PHF6 (11.5%) in T cell ALL (T-ALL). These mutant genes are involved in key molecular processes, including the Ras pathway, the Notch pathway, epigenetic modification, and cell-cycle regulation. Strikingly, more than 50% of mutations occurred in the high-hyperdiploid (HeH) ALL existed in Ras pathway, especially FLT3 (20%). We also found that the epigenetic regulator gene KMT2D, which is frequently mutated in ALL, may be involved in driving leukemia transformation, as evidenced by an in vitro functional assay. CONCLUSION: Overall, this study provides further insights into the genetic basis of ALL and shows that Ras mutations are predominant in childhood ALL, especially in the high-hyperdiploid subtype in our research.
format Online
Article
Text
id pubmed-7068927
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-70689272020-03-18 Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing Zhang, Honghong Wang, Hongsheng Qian, Xiaowen Gao, Shuai Xia, Jieqi Liu, Junwen Cheng, Yanqin Man, Jie Zhai, Xiaowen BMC Cancer Research Article BACKGROUND: Acute lymphoblastic leukemia (ALL), the most common childhood malignancy, is characterized by recurring structural chromosomal alterations and genetic alterations, whose detection is critical in diagnosis, risk stratification and prognostication. However, the genetic mechanisms that give rise to ALL remain poorly understood. METHODS: Using next-generation sequencing (NGS) in matched germline and tumor samples from 140 pediatric Chinese patients with ALL, we landscaped the gene mutations and estimated the mutation frequencies in this disease. RESULTS: Our results showed that the top driver oncogenes having a mutation prevalence over 5% in childhood ALL included KRAS (8.76%), NRAS (6.4%), FLT3 (5.7%) and KMT2D (5.0%). While the most frequently mutated genes were KRAS, NRAS and FLT3 in B cell ALL (B-ALL), the most common mutations were enriched in NOTCH1 (23.1%), FBXW7 (23.1%) and PHF6 (11.5%) in T cell ALL (T-ALL). These mutant genes are involved in key molecular processes, including the Ras pathway, the Notch pathway, epigenetic modification, and cell-cycle regulation. Strikingly, more than 50% of mutations occurred in the high-hyperdiploid (HeH) ALL existed in Ras pathway, especially FLT3 (20%). We also found that the epigenetic regulator gene KMT2D, which is frequently mutated in ALL, may be involved in driving leukemia transformation, as evidenced by an in vitro functional assay. CONCLUSION: Overall, this study provides further insights into the genetic basis of ALL and shows that Ras mutations are predominant in childhood ALL, especially in the high-hyperdiploid subtype in our research. BioMed Central 2020-03-12 /pmc/articles/PMC7068927/ /pubmed/32164600 http://dx.doi.org/10.1186/s12885-020-6709-7 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
Zhang, Honghong
Wang, Hongsheng
Qian, Xiaowen
Gao, Shuai
Xia, Jieqi
Liu, Junwen
Cheng, Yanqin
Man, Jie
Zhai, Xiaowen
Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title_full Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title_fullStr Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title_full_unstemmed Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title_short Genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in China using exon sequencing
title_sort genetic mutational analysis of pediatric acute lymphoblastic leukemia from a single center in china using exon sequencing
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7068927/
https://www.ncbi.nlm.nih.gov/pubmed/32164600
http://dx.doi.org/10.1186/s12885-020-6709-7
work_keys_str_mv AT zhanghonghong geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT wanghongsheng geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT qianxiaowen geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT gaoshuai geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT xiajieqi geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT liujunwen geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT chengyanqin geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT manjie geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing
AT zhaixiaowen geneticmutationalanalysisofpediatricacutelymphoblasticleukemiafromasinglecenterinchinausingexonsequencing