Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
N Engl J Med ; 365(15): 1384-95, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21995386

ABSTRACT

BACKGROUND: Myelodysplastic syndromes are a diverse and common group of chronic hematologic cancers. The identification of new genetic lesions could facilitate new diagnostic and therapeutic strategies. METHODS: We used massively parallel sequencing technology to identify somatically acquired point mutations across all protein-coding exons in the genome in 9 patients with low-grade myelodysplasia. Targeted resequencing of the gene encoding RNA splicing factor 3B, subunit 1 (SF3B1), was also performed in a cohort of 2087 patients with myeloid or other cancers. RESULTS: We identified 64 point mutations in the 9 patients. Recurrent somatically acquired mutations were identified in SF3B1. Follow-up revealed SF3B1 mutations in 72 of 354 patients (20%) with myelodysplastic syndromes, with particularly high frequency among patients whose disease was characterized by ring sideroblasts (53 of 82 [65%]). The gene was also mutated in 1 to 5% of patients with a variety of other tumor types. The observed mutations were less deleterious than was expected on the basis of chance, suggesting that the mutated protein retains structural integrity with altered function. SF3B1 mutations were associated with down-regulation of key gene networks, including core mitochondrial pathways. Clinically, patients with SF3B1 mutations had fewer cytopenias and longer event-free survival than patients without SF3B1 mutations. CONCLUSIONS: Mutations in SF3B1 implicate abnormalities of messenger RNA splicing in the pathogenesis of myelodysplastic syndromes. (Funded by the Wellcome Trust and others.).


Subject(s)
Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Point Mutation , Ribonucleoprotein, U2 Small Nuclear/genetics , Erythrocytes/pathology , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Phenotype , RNA Splicing Factors
2.
Leukemia ; 30(12): 2322-2331, 2016 12.
Article in English | MEDLINE | ID: mdl-27211273

ABSTRACT

The splicing factor SF3B1 is the most frequently mutated gene in myelodysplastic syndromes (MDS), and is strongly associated with the presence of ring sideroblasts (RS). We have performed a systematic analysis of cryptic splicing abnormalities from RNA sequencing data on hematopoietic stem cells (HSCs) of SF3B1-mutant MDS cases with RS. Aberrant splicing events in many downstream target genes were identified and cryptic 3' splice site usage was a frequent event in SF3B1-mutant MDS. The iron transporter ABCB7 is a well-recognized candidate gene showing marked downregulation in MDS with RS. Our analysis unveiled aberrant ABCB7 splicing, due to usage of an alternative 3' splice site in MDS patient samples, giving rise to a premature termination codon in the ABCB7 mRNA. Treatment of cultured SF3B1-mutant MDS erythroblasts and a CRISPR/Cas9-generated SF3B1-mutant cell line with the nonsense-mediated decay (NMD) inhibitor cycloheximide showed that the aberrantly spliced ABCB7 transcript is targeted by NMD. We describe cryptic splicing events in the HSCs of SF3B1-mutant MDS, and our data support a model in which NMD-induced downregulation of the iron exporter ABCB7 mRNA transcript resulting from aberrant splicing caused by mutant SF3B1 underlies the increased mitochondrial iron accumulation found in MDS patients with RS.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Base Sequence , Cycloheximide/pharmacology , Hematopoietic Stem Cells/metabolism , Humans , Iron/metabolism , Mitochondria/metabolism , RNA Splicing , Tumor Cells, Cultured
3.
Leukemia ; 29(5): 1092-103, 2015 May.
Article in English | MEDLINE | ID: mdl-25428262

ABSTRACT

The splicing factor SF3B1 is the most commonly mutated gene in the myelodysplastic syndrome (MDS), particularly in patients with refractory anemia with ring sideroblasts (RARS). We investigated the functional effects of SF3B1 disruption in myeloid cell lines: SF3B1 knockdown resulted in growth inhibition, cell cycle arrest and impaired erythroid differentiation and deregulation of many genes and pathways, including cell cycle regulation and RNA processing. MDS is a disorder of the hematopoietic stem cell and we thus studied the transcriptome of CD34(+) cells from MDS patients with SF3B1 mutations using RNA sequencing. Genes significantly differentially expressed at the transcript and/or exon level in SF3B1 mutant compared with wild-type cases include genes that are involved in MDS pathogenesis (ASXL1 and CBL), iron homeostasis and mitochondrial metabolism (ALAS2, ABCB7 and SLC25A37) and RNA splicing/processing (PRPF8 and HNRNPD). Many genes regulated by a DNA damage-induced BRCA1-BCLAF1-SF3B1 protein complex showed differential expression/splicing in SF3B1 mutant cases. This is the first study to determine the target genes of SF3B1 mutation in MDS CD34(+) cells. Our data indicate that SF3B1 has a critical role in MDS by affecting the expression and splicing of genes involved in specific cellular processes/pathways, many of which are relevant to the known RARS pathophysiology, suggesting a causal link.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Stem Cells/cytology , Alternative Splicing , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Antigens, CD34/metabolism , Cell Cycle , Cell Differentiation , Cell Proliferation , Exons , Female , Gene Expression Profiling , Gene Knockdown Techniques , Heterozygote , Homeostasis , Humans , K562 Cells , Male , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , Point Mutation , RNA/genetics , RNA Splicing , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/metabolism , Sequence Analysis, RNA
4.
Neurobiol Aging ; 22(4): 563-8, 2001.
Article in English | MEDLINE | ID: mdl-11445256

ABSTRACT

In the present study we analysed the genotype of HFE, the gene involved in hemochromatosis, in 107 patients with sporadic late-onset AD and in 99 age-matched non-demented controls. We observed that patients carrying the mutant HFE-H63D allele had a mean age at onset of 71.7 +/- 6.0 years versus 76.6 +/- 5.8 years of those who were homozygous for the wild-type allele (p = 0.001). The frequency of the HFE-H63D mutation was highest (0.22) in the patients aged <70 years at the time of disease onset, whereas it was 0.12 in those with disease onset at an age of 70-80 years, and 0.04 in those aged more than 80 years. The APOE genotype did not significantly modify the effect of HFE on age at onset. We conclude that mild disturbances of iron homeostasis associated with a common genetic determinant may interact with other pathogenic mechanisms involved in AD. HFE mutations may anticipate AD clinical presentation in susceptible individuals.


Subject(s)
Alzheimer Disease/genetics , HLA Antigens/genetics , Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins , Age of Onset , Aged , Aged, 80 and over , Apolipoprotein E4 , Apolipoproteins E/genetics , Female , Gene Frequency , Genotype , Hemochromatosis Protein , Humans , Male , Middle Aged , Mutation , Risk Factors
6.
Leukemia ; 24(6): 1139-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20410925

ABSTRACT

We have undertaken a genome-wide single nucleotide polymorphism (SNP) array analysis of 41 chronic myeloid leukemia (CML) patients. In total, 44 regions of uniparental disomy (UPD) >3 Mb were identified in 24 of 32 patients in chronic phase (CP), and 21 regions of UPD >3 Mb were identified in 13 of 21 patients in blast crisis (BC). Chromosome 8 had the highest frequency of UPD regions in both CP and BC samples. Eight recurrent regions of UPD were observed among the 41 patients, with chromosome 8 showing the highest frequency. Ten regions of copy number change (CNC) >3 Mb were observed in 4 of 21 patients in BC, whereas none were observed in CP. We have identified several recurrent regions of UPD and CNC in CML that may be of pathogenetic importance. Overrepresentation of genomic aberrations (UPD and copy number gain) mapping to chromosome 8 was observed. Selected candidate genes mapping within the aberrant genomic regions were sequenced and mutation of the TP53 gene was observed in one case in BC and of the ASXL1 gene in 6 of 41 cases in CP or BC. Mutation of ASXL1 represents an important new molecular abnormality in CML.


Subject(s)
Blast Crisis/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Repressor Proteins/genetics , Uniparental Disomy/genetics , Disease Progression , Gene Dosage , Genome, Human , Humans , Karyotyping , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Oligonucleotide Array Sequence Analysis , Tumor Suppressor Protein p53/genetics
7.
Leukemia ; 24(4): 756-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20220779

ABSTRACT

To gain insight into the molecular pathogenesis of the myelodysplastic syndromes (MDS), we performed global gene expression profiling and pathway analysis on the hematopoietic stem cells (HSC) of 183 MDS patients as compared with the HSC of 17 healthy controls. The most significantly deregulated pathways in MDS include interferon signaling, thrombopoietin signaling and the Wnt pathways. Among the most significantly deregulated gene pathways in early MDS are immunodeficiency, apoptosis and chemokine signaling, whereas advanced MDS is characterized by deregulation of DNA damage response and checkpoint pathways. We have identified distinct gene expression profiles and deregulated gene pathways in patients with del(5q), trisomy 8 or -7/del(7q). Patients with trisomy 8 are characterized by deregulation of pathways involved in the immune response, patients with -7/del(7q) by pathways involved in cell survival, whereas patients with del(5q) show deregulation of integrin signaling and cell cycle regulation pathways. This is the first study to determine deregulated gene pathways and ontology groups in the HSC of a large group of MDS patients. The deregulated pathways identified are likely to be critical to the MDS HSC phenotype and give new insights into the molecular pathogenesis of this disorder, thereby providing new targets for therapeutic intervention.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Myelodysplastic Syndromes/genetics , Signal Transduction , Biomarkers, Tumor/metabolism , Case-Control Studies , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 8/genetics , Hematopoietic Stem Cells/pathology , Humans , Myelodysplastic Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trisomy
SELECTION OF CITATIONS
SEARCH DETAIL