Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nat Commun ; 14(1): 1658, 2023 03 25.
Article in English | MEDLINE | ID: mdl-36966135

ABSTRACT

High hyperdiploid acute lymphoblastic leukemia (HeH ALL), one of the most common childhood malignancies, is driven by nonrandom aneuploidy (abnormal chromosome numbers) mainly comprising chromosomal gains. In this study, we investigate how aneuploidy in HeH ALL arises. Single cell whole genome sequencing of 2847 cells from nine primary cases and one normal bone marrow reveals that HeH ALL generally display low chromosomal heterogeneity, indicating that they are not characterized by chromosomal instability and showing that aneuploidy-driven malignancies are not necessarily chromosomally heterogeneous. Furthermore, most chromosomal gains are present in all leukemic cells, suggesting that they arose early during leukemogenesis. Copy number data from 577 primary cases reveals selective pressures that were used for in silico modeling of aneuploidy development. This shows that the aneuploidy in HeH ALL likely arises by an initial tripolar mitosis in a diploid cell followed by clonal evolution, in line with a punctuated evolution model.


Subject(s)
Aneuploidy , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Chromosome Aberrations , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Diploidy , Chromosomal Instability
2.
Blood ; 136(8): 946-956, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32384149

ABSTRACT

Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene in 13q12.2 are among the most common driver events in acute leukemia, leading to increased cell proliferation and survival through activation of the phosphatidylinositol 3-kinase/AKT-, RAS/MAPK-, and STAT5-signaling pathways. In this study, we examine the pathogenetic impact of somatic hemizygous 13q12.2 microdeletions in B-cell precursor (BCP) acute lymphoblastic leukemia (ALL) using 5 different patient cohorts (in total including 1418 cases). The 13q12.2 deletions occur immediately 5' of FLT3 and involve the PAN3 locus. By detailed analysis of the 13q12.2 segment, we show that the deletions lead to loss of a topologically associating domain border and an enhancer of FLT3. This results in increased cis interactions between the FLT3 promoter and another enhancer located distally to the deletion breakpoints, with subsequent allele-specific upregulation of FLT3 expression, expected to lead to ligand-independent activation of the receptor and downstream signaling. The 13q12.2 deletions are highly enriched in the high-hyperdiploid BCP ALL subtype (frequency 3.9% vs 0.5% in other BCP ALL) and in cases that subsequently relapsed. Taken together, our study describes a novel mechanism of FLT3 involvement in leukemogenesis by upregulation via chromatin remodeling and enhancer hijacking. These data further emphasize the role of FLT3 as a driver gene in BCP ALL.


Subject(s)
Chromosome Disorders/genetics , Enhancer Elements, Genetic/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , fms-Like Tyrosine Kinase 3/genetics , Cell Line , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Chromosome Deletion , Chromosome Disorders/complications , Chromosomes, Human, Pair 13/genetics , Cohort Studies , DNA Copy Number Variations/genetics , Gene Expression Regulation, Leukemic , Humans , Microarray Analysis , Polymorphism, Single Nucleotide , RNA-Seq , Up-Regulation/genetics , Whole Genome Sequencing
3.
Nat Commun ; 10(1): 1519, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944321

ABSTRACT

Hyperdiploidy, i.e. gain of whole chromosomes, is one of the most common genetic features of childhood acute lymphoblastic leukemia (ALL), but its pathogenetic impact is poorly understood. Here, we report a proteogenomic analysis on matched datasets from genomic profiling, RNA-sequencing, and mass spectrometry-based analysis of >8,000 genes and proteins as well as Hi-C of primary patient samples from hyperdiploid and ETV6/RUNX1-positive pediatric ALL. We show that CTCF and cohesin, which are master regulators of chromatin architecture, display low expression in hyperdiploid ALL. In line with this, a general genome-wide dysregulation of gene expression in relation to topologically associating domain (TAD) borders were seen in the hyperdiploid group. Furthermore, Hi-C of a limited number of hyperdiploid childhood ALL cases revealed that 2/4 cases displayed a clear loss of TAD boundary strength and 3/4 showed reduced insulation at TAD borders, with putative leukemogenic effects.


Subject(s)
Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription, Genetic , Adolescent , Aneuploidy , CCCTC-Binding Factor/genetics , Cell Cycle Proteins/genetics , Child , Child, Preschool , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Aberrations , Core Binding Factor Alpha 2 Subunit/genetics , Female , Gene Dosage , Gene Expression Profiling , Genome, Human , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Male , Proteogenomics/methods , Proteome/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Sequence Analysis, RNA , Cohesins , ETS Translocation Variant 6 Protein
4.
Cancers (Basel) ; 11(3)2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30909364

ABSTRACT

Anaplastic thyroid cancer (ATC) is a rare and extremely malignant tumor with no available cure. The genetic landscape of this malignancy has not yet been fully explored. In this study, we performed whole exome sequencing and the RNA-sequencing of fourteen cases of ATC to delineate copy number changes, fusion gene events, and somatic mutations. A high frequency of genomic amplifications was seen, including 29% of cases having amplification of CCNE1 and 9% of CDK6; these events may be targetable by cyclin dependent kinase (CDK) inhibition. Furthermore, 9% harbored amplification of TWIST1, which is also a potentially targetable lesion. A total of 21 fusion genes in five cases were seen, none of which were recurrent. Frequent mutations included TP53 (55%), the TERT promoter (36%), and ATM (27%). Analyses of mutational signatures showed an involvement of processes that are associated with normal aging, defective DNA mismatch repair, activation induced cytidine deaminase (AID)/apolipoprotein B editing complex (APOBEC) activity, failure of DNA double-strand break repair, and tobacco exposure. Taken together, our results shed new light on the tumorigenesis of ATC and show that a relatively large proportion (36%) of ATCs harbor genetic events that make them candidates for novel therapeutic approaches. When considering that ATC today has a mortality rate of close to 100%, this is highly relevant from a clinical perspective.

6.
Endocr Relat Cancer ; 24(5): 209-220, 2017 05.
Article in English | MEDLINE | ID: mdl-28235956

ABSTRACT

Anaplastic thyroid cancer (ATC) is a highly malignant disease with a very short median survival time. Few studies have addressed the underlying somatic mutations, and the genomic landscape of ATC thus remains largely unknown. In the present study, we have ascertained copy number aberrations, gene fusions, gene expression patterns, and mutations in early-passage cells from ten newly established ATC cell lines using single nucleotide polymorphism (SNP) array analysis, RNA sequencing and whole exome sequencing. The ATC cell line genomes were highly complex and displayed signs of replicative stress and genomic instability, including massive aneuploidy and frequent breakpoints in the centromeric regions and in fragile sites. Loss of heterozygosity involving whole chromosomes was common, but there were no signs of previous near-haploidisation events or chromothripsis. A total of 21 fusion genes were detected, including six predicted in-frame fusions; none were recurrent. Global gene expression analysis showed 661 genes to be differentially expressed between ATC and papillary thyroid cancer cell lines, with pathway enrichment analyses showing downregulation of TP53 signalling as well as cell adhesion molecules in ATC. Besides previously known driver events, such as mutations in BRAF, NRAS, TP53 and the TERT promoter, we identified PTPRD and NEGR1 as putative novel target genes in ATC, based on deletions in six and four cell lines, respectively; the latter gene also carried a somatic mutation in one cell line. Taken together, our data provide novel insights into the tumourigenesis of ATC and may be used to identify new therapeutic targets.


Subject(s)
Genetic Heterogeneity , Genomic Instability , Molecular Targeted Therapy/methods , Thyroid Carcinoma, Anaplastic/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL