Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Clin Oncol ; 31(28): 3557-64, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24002510

ABSTRACT

PURPOSE: The diagnosis of patients with myelodysplastic syndromes (MDS) is largely dependent on morphologic examination of bone marrow aspirates. Several criteria that form the basis of the classifications and scoring systems most commonly used in clinical practice are affected by operator-dependent variation. To identify standardized molecular markers that would allow prediction of prognosis, we have used gene expression profiling (GEP) data on CD34+ cells from patients with MDS to determine the relationship between gene expression levels and prognosis. PATIENTS AND METHODS: GEP data on CD34+ cells from 125 patients with MDS with a minimum 12-month follow-up since date of bone marrow sample collection were included in this study. Supervised principal components and lasso penalized Cox proportional hazards regression (Coxnet) were used for the analysis. RESULTS: We identified several genes, the expression of which was significantly associated with survival of patients with MDS, including LEF1, CDH1, WT1, and MN1. The Coxnet predictor, based on expression data on 20 genes, outperformed other predictors, including one that additionally used clinical information. Our Coxnet gene signature based on CD34+ cells significantly identified a separation of patients with good or bad prognosis in an independent GEP data set based on unsorted bone marrow mononuclear cells, demonstrating that our signature is robust and may be applicable to bone marrow cells without the need to isolate CD34+ cells. CONCLUSION: We present a new, valuable GEP-based signature for assessing prognosis in MDS. GEP-based signatures correlating with clinical outcome may significantly contribute to a refined risk classification of MDS.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Myelodysplastic Syndromes/diagnosis , Neoplastic Stem Cells/metabolism , Aged , Antigens, CD34/metabolism , Female , Follow-Up Studies , Hematopoietic Stem Cells/pathology , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Prognosis , Survival Rate
2.
Haematologica ; 98(12): 1856-64, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23831921

ABSTRACT

Interstitial deletion of chromosome 5q is the most common chromosomal abnormality in myelodysplastic syndromes. The catalogue of genes involved in the molecular pathogenesis of myelodysplastic syndromes is rapidly expanding and next-generation sequencing technology allows detection of these mutations at great depth. Here we describe the design, validation and application of a targeted next-generation sequencing approach to simultaneously screen 25 genes mutated in myeloid malignancies. We used this method alongside single nucleotide polymorphism-array technology to characterize the mutational and cytogenetic profile of 43 cases of early or advanced del(5q) myelodysplastic syndromes. A total of 29 mutations were detected in our cohort. Overall, 45% of early and 66.7% of advanced cases had at least one mutation. Genes with the highest mutation frequency among advanced cases were TP53 and ASXL1 (25% of patients each). These showed a lower mutation frequency in cases of 5q- syndrome (4.5% and 13.6%, respectively), suggesting a role in disease progression in del(5q) myelodysplastic syndromes. Fifty-two percent of mutations identified were in genes involved in epigenetic regulation (ASXL1, TET2, DNMT3A and JAK2). Six mutations had allele frequencies <20%, likely below the detection limit of traditional sequencing methods. Genomic array data showed that cases of advanced del(5q) myelodysplastic syndrome had a complex background of cytogenetic aberrations, often encompassing genes involved in myeloid disorders. Our study is the first to investigate the molecular pathogenesis of early and advanced del(5q) myelodysplastic syndromes using next-generation sequencing technology on a large panel of genes frequently mutated in myeloid malignancies, further illuminating the molecular landscape of del(5q) myelodysplastic syndromes.


Subject(s)
Anemia, Macrocytic/genetics , Gene Targeting/methods , Mutation/genetics , Myelodysplastic Syndromes/genetics , Sequence Analysis, DNA/methods , Adult , Aged , Aged, 80 and over , Anemia, Macrocytic/diagnosis , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Cohort Studies , Female , Gene Frequency/genetics , Humans , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Polymorphism, Single Nucleotide/genetics , Young Adult
3.
Br J Haematol ; 160(6): 842-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23294243

ABSTRACT

The ASXL1 gene encodes a chromatin-binding protein involved in epigenetic regulation in haematopoietic cells. Loss-of-function ASXL1 mutations occur in patients with a range of myeloid malignancies and are associated with adverse outcome. We have used lentiviral-based shRNA technology to investigate the effects of ASXL1 silencing on cell proliferation, apoptosis, myeloid differentiation and global gene expression in human CD34(+) cells differentiated along the myeloid lineage in vitro. ASXL1-deficient cells showed a significant decrease in the generation of CD11b(+) and CD15(+) cells, implicating impaired granulomonocytic differentiation. Furthermore, colony-forming assays showed a significant increase in the number of multipotent mixed lineage colony-forming unit (CFU-GEMM) colonies and a significant decrease in the numbers of granulocyte-macrophage CFU (CFU-GM) and granulocyte CFU (CFU-G) colonies in ASXL1-deficient cells. Our data suggests that ASXL1 knockdown perturbs human granulomonocytic differentiation. Gene expression profiling identified many deregulated genes in the ASXL1-deficient cells differentiated along the granulomonocytic lineage, and pathway analysis showed that the most significantly deregulated pathway was the LXR/RXR activation pathway. ASXL1 may play a key role in recruiting the polycomb repressor complex 2 (PRC2) to specific loci, and we found over-representation of PRC2 targets among the deregulated genes in ASXL1-deficient cells. These findings shed light on the functional role of ASXL1 in human myeloid differentiation.


Subject(s)
Antigens, CD34/biosynthesis , Myeloid Cells/physiology , Repressor Proteins/genetics , Stem Cells/physiology , Case-Control Studies , Cell Culture Techniques , Cell Differentiation/genetics , Cell Growth Processes/genetics , Cell Lineage , Gene Silencing , Humans , K562 Cells , Myeloid Cells/cytology , Myeloid Cells/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome
4.
Adv Biol Regul ; 53(1): 8-17, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23031788

ABSTRACT

Patients with the 5q- syndrome and Diamond-Blackfan anemia (DBA) suffer from a severe macrocytic anemia. The 5q- syndrome and DBA are disorders of aberrant ribosome biogenesis (ribosomopathies) and haploinsufficiency of the ribosomal protein genes RPS14 and RPS19, respectively, underlies the anemia found in these disorders. Erythroblasts obtained from patients with the 5q- syndrome and DBA show impaired mRNA translation and this defect in translation may represent a potential therapeutic target in these ribosomopathies. There are some indications that the amino acid l-leucine, a translation enhancer, may have some efficacy in this group of disorders. Recent studies have shown that l-leucine treatment of zebrafish and murine models of the 5q- syndrome and DBA results in a marked improvement in the anemia. l-leucine treatment of RPS14-deficient and RPS19-deficient erythroblasts and erythroblasts from patients with the 5q- syndrome has been shown to result in an increase in cell proliferation, erythroid differentiation and mRNA translation in culture. l-leucine has been shown to improve hemoglobin levels and transfusion independence in a patient with DBA. l-leucine activates the mTOR (mammalian target of rapamycin) signaling pathway that controls cell growth and mRNA translation. There is evidence to suggest that the promotion of translation via the mTOR pathway by l-leucine is the mechanism that underlies the enhanced erythroid progenitor cell growth and differentiation observed in animal and cellular models of the 5q- syndrome and DBA treated with this amino acid. These data support the rationale for clinical trials of l-leucine as a therapeutic agent for the 5q- syndrome and DBA.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , Anemia, Macrocytic/metabolism , Leucine/pharmacology , Ribosomes/drug effects , Signal Transduction/drug effects , Anemia, Diamond-Blackfan/drug therapy , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Anemia, Macrocytic/drug therapy , Anemia, Macrocytic/genetics , Anemia, Macrocytic/pathology , Animals , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythroblasts/pathology , Gene Expression Regulation/drug effects , Haploinsufficiency , Humans , Leucine/metabolism , Protein Biosynthesis , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 7(8): e42334, 2012.
Article in English | MEDLINE | ID: mdl-22912701

ABSTRACT

Acute myeloid leukemia patients with normal cytogenetics (CN-AML) account for almost half of AML cases. We aimed to study the frequency and relationship of a wide range of genes previously reported as mutated in AML (ASXL1, NPM1, FLT3, TET2, IDH1/2, RUNX1, DNMT3A, NRAS, JAK2, WT1, CBL, SF3B1, TP53, KRAS and MPL) in a series of 84 CN-AML cases. The most frequently mutated genes in primary cases were NPM1 (60.8%) and FLT3 (50.0%), and in secondary cases ASXL1 (48.5%) and TET2 (30.3%). We showed that 85% of CN-AML patients have mutations in at least one of ASXL1, NPM1, FLT3, TET2, IDH1/2 and/or RUNX1. Serial samples from 19 MDS/CMML cases that progressed to AML were analyzed for ASXL1/TET2/IDH1/2 mutations; seventeen cases presented mutations of at least one of these genes. However, there was no consistent pattern in mutation acquisition during disease progression. This report concerns the analysis of the largest number of gene mutations in CN-AML studied to date, and provides insight into the mutational profile of CN-AML.


Subject(s)
Cytogenetic Analysis , DNA Mutational Analysis , Genes, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Karyotype , Leukemia, Myeloid, Acute/diagnosis , Male , Middle Aged , Neoplasm Metastasis , Nucleophosmin , Prognosis , Young Adult
6.
Curr Pharm Des ; 18(22): 3180-3, 2012.
Article in English | MEDLINE | ID: mdl-22571696

ABSTRACT

In recent years we have gained great insight into the molecular pathogenesis of the 5q- syndrome, the most distinct of all the myelodysplastic syndromes. It is now recognized that p53 activation, caused by haploinsufficiency for the ribosomal gene RPS14 (mapping to the commonly deleted region), is the probable cause of the erythroid defect in the 5q- syndrome. A mouse model of the human 5q- syndrome has been generated by large-scale deletion of the Cd74-Nid67 interval (containing Rps14) and the crossing of these '5q- mice' with p53-deficient mice ameliorated the erythroid progenitor defect. Recent evidence suggests that haploinsufficiency of the microRNA genes miR-145 and miR-146a may contribute to the thrombocytosis seen in the 5q- syndrome. Emerging data shows that p53 mutation may play a role in disease progression.


Subject(s)
Anemia, Macrocytic/physiopathology , Myelodysplastic Syndromes/physiopathology , Tumor Suppressor Protein p53/genetics , Anemia, Macrocytic/genetics , Animals , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Disease Models, Animal , Disease Progression , Erythroid Precursor Cells/pathology , Haploinsufficiency , Humans , Mice , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Ribosomal Proteins/genetics
7.
PLoS One ; 7(2): e31605, 2012.
Article in English | MEDLINE | ID: mdl-22328940

ABSTRACT

Chronic myelomonocytic leukemia (CMML) has recently been associated with a high incidence of diverse mutations in genes such as TET2 or EZH2 that are implicated in epigenetic mechanisms. We have performed genome-wide DNA methylation arrays and mutational analysis of TET2, IDH1, IDH2, EZH2 and JAK2 in a group of 24 patients with CMML. 249 genes were differentially methylated between CMML patients and controls. Using Ingenuity pathway analysis, we identified enrichment in a gene network centered around PLC, JNK and ERK suggesting that these pathways, whose deregulation has been recently described in CMML, are affected by epigenetic mechanisms. Mutations of TET2, JAK2 and EZH2 were found in 15 patients (65%), 4 patients (17%) and 1 patient (4%) respectively while no mutations in the IDH1 and IDH2 genes were identified. Interestingly, patients with wild type TET2 clustered separately from patients with TET2 mutations, showed a higher degree of hypermethylation and were associated with higher risk karyotypes. Our results demonstrate the presence of aberrant DNA methylation in CMML and identifies TET2 mutant CMML as a biologically distinct disease subtype with a different epigenetic profile.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Proto-Oncogene Proteins/genetics , 5-Methylcytosine , Computational Biology , Cytosine/analogs & derivatives , DNA Methylation/genetics , Dioxygenases , Enhancer of Zeste Homolog 2 Protein , Humans , Isocitrate Dehydrogenase/genetics , Janus Kinase 2/genetics , Mutation , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2 , Transcription Factors/genetics
9.
Blood ; 118(24): 6239-46, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21998214

ABSTRACT

In a previous study, we identified somatic mutations of SF3B1, a gene encoding a core component of RNA splicing machinery, in patients with myelodysplastic syndrome (MDS). Here, we define the clinical significance of these mutations in MDS and myelodysplastic/myeloproliferative neoplasms (MDS/MPN). The coding exons of SF3B1 were screened using massively parallel pyrosequencing in patients with MDS, MDS/MPN, or acute myeloid leukemia (AML) evolving from MDS. Somatic mutations of SF3B1 were found in 150 of 533 (28.1%) patients with MDS, 16 of 83 (19.3%) with MDS/MPN, and 2 of 38 (5.3%) with AML. There was a significant association of SF3B1 mutations with the presence of ring sideroblasts (P < .001) and of mutant allele burden with their proportion (P = .002). The mutant gene had a positive predictive value for ring sideroblasts of 97.7% (95% confidence interval, 93.5%-99.5%). In multivariate analysis including established risk factors, SF3B1 mutations were found to be independently associated with better overall survival (hazard ratio = 0.15, P = .025) and lower risk of evolution into AML (hazard ratio = 0.33, P = .049). The close association between SF3B1 mutations and disease phenotype with ring sideroblasts across MDS and MDS/MPN is consistent with a causal relationship. Furthermore, SF3B1 mutations are independent predictors of favorable clinical outcome, and their incorporation into stratification systems might improve risk assessment in MDS.


Subject(s)
Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Myelodysplastic-Myeloproliferative Diseases/genetics , Myelodysplastic-Myeloproliferative Diseases/physiopathology , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Aged , Alleles , Codon , DNA Mutational Analysis , Erythroblasts/pathology , Female , Follow-Up Studies , Genetic Association Studies , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/pathology , Prognosis , RNA Splicing Factors , Sex Characteristics , Survival Analysis
10.
PLoS One ; 6(6): e20607, 2011.
Article in English | MEDLINE | ID: mdl-21694761

ABSTRACT

Leukaemia is often associated with genetic alterations such as translocations, amplifications and deletions, and recurrent chromosome abnormalities are used as markers of diagnostic and prognostic relevance. However, a proportion of acute myeloid leukaemia (AML) cases have an apparently normal karyotype despite comprehensive cytogenetic analysis. Based on conventional cytogenetic analysis of banded chromosomes, we selected a series of 23 paediatric patients with acute myeloid leukaemia and performed whole genome array comparative genome hybridization (aCGH) using DNA samples derived from the same patients. Imbalances involving large chromosomal regions or entire chromosomes were detected by aCGH in seven of the patients studied. Results were validated by fluorescence in situ hybridization (FISH) to both interphase nuclei and metaphase chromosomes using appropriate bacterial artificial chromosome (BAC) probes. The majority of these copy number alterations (CNAs) were confirmed by FISH and found to localize to the interphase rather than metaphase nuclei. Furthermore, the proliferative states of the cells analyzed by FISH were tested by immunofluorescence using an antibody against the proliferation marker pKi67. Interestingly, these experiments showed that, in the vast majority of cases, the changes appeared to be confined to interphase nuclei in a non-proliferative status.


Subject(s)
Genome, Human/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Adult , Cell Nucleus/genetics , Cell Proliferation , Child , Chromosome Aberrations , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Female , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotyping , Male , Reproducibility of Results
11.
Biol Direct ; 6: 23, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21592325

ABSTRACT

BACKGROUND: MicroRNAs are small RNA species that regulate gene expression post-transcriptionally and are aberrantly expressed in many cancers including hematological malignancies. However, the role of microRNAs in the pathogenesis of multiple myeloma (MM) is only poorly understood. We therefore used microarray analysis to elucidate the complete miRNome (miRBase version 13.0) of purified tumor (CD138+) cells from 33 patients with MM, 5 patients with monoclonal gammopathy of undetermined significance (MGUS) and 9 controls. RESULTS: Unsupervised cluster analysis revealed that MM and MGUS samples have a distinct microRNA expression profile from control CD138+ cells. The majority of microRNAs aberrantly expressed in MM (109/129) were up-regulated. A comparison of these microRNAs with those aberrantly expressed in other B-cell and T-cell malignancies revealed a surprising degree of similarity (~40%) suggesting the existence of a common lymphoma microRNA signature. We identified 39 microRNAs associated with the pre-malignant condition MGUS. Twenty-three (59%) of these were also aberrantly expressed in MM suggesting common microRNA expression events in MM progression. MM is characterized by multiple chromosomal abnormalities of varying prognostic significance. We identified specific microRNA signatures associated with the most common IgH translocations (t(4;14) and t(11;14)) and del(13q). Expression levels of these microRNAs were distinct between the genetic subtypes (by cluster analysis) and correctly predicted these abnormalities in > 85% of cases using the support vector machine algorithm. Additionally, we identified microRNAs associated with light chain only myeloma, as well as IgG and IgA-type MM. Finally, we identified 32 microRNAs associated with event-free survival (EFS) in MM, ten of which were significant by univariate (logrank) survival analysis. CONCLUSIONS: In summary, this work has identified aberrantly expressed microRNAs associated with the diagnosis, pathogenesis and prognosis of MM, data which will prove an invaluable resource for understanding the role of microRNAs in this devastating disease.


Subject(s)
MicroRNAs/genetics , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Neoplasms, Plasma Cell/genetics , Adult , Aged , Aged, 80 and over , Cluster Analysis , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/classification , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/classification , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/classification , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Neoplasms, Plasma Cell/classification , Neoplasms, Plasma Cell/diagnosis , Neoplasms, Plasma Cell/pathology , Oligonucleotide Array Sequence Analysis , Prognosis , Translocation, Genetic , Up-Regulation
13.
Blood ; 116(26): 5803-11, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20733155

ABSTRACT

The 5q- syndrome is the most distinct of all the myelodysplastic syndromes with a clear genotype/phenotype relationship. The significant progress made during recent years has been based on the determination of the commonly deleted region and the demonstration of haploinsufficiency for the ribosomal gene RPS14. The functional screening of all the genes in the commonly deleted region determined that RPS14 haploinsufficiency is the probable cause of the erythroid defect in the 5q- syndrome. A mouse model of the human 5q- syndrome has now been created by chromosomal engineering involving a large-scale deletion of the Cd74-Nid67 interval (containing RPS14). A variety of lines of evidence support the model of ribosomal deficiency causing p53 activation and defective erythropoiesis, including most notably the crossing of the "5q- mice" with p53-deficient mice, thereby ameliorating the erythroid progenitor defect. Emerging evidence supports the notion that the p53 activation observed in the mouse model may also apply to the human 5q- syndrome. Other mouse modeling data suggest that haploinsufficiency of the microRNA genes miR-145 and miR-146a may contribute to the thrombocytosis seen in the 5q- syndrome. Lenalidomide has become an established therapy for the 5q- syndrome, although its precise mode of action remains uncertain.


Subject(s)
MicroRNAs/genetics , Anemia, Macrocytic/genetics , Animals , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Cri-du-Chat Syndrome/genetics , Haploinsufficiency , Humans , Mice , Trisomy/genetics
14.
Blood ; 116(7): 1105-13, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20448109

ABSTRACT

MicroRNAs are commonly aberrantly expressed in many cancers. Very little is known of their role in T-cell lymphoma, however. We therefore elucidated the complete miRNome of purified T cells from 21 patients diagnosed with Sézary Syndrome (SzS), a rare aggressive primary cutaneous T-cell (CD4(+)) lymphoma. Unsupervised cluster analysis of microarray data revealed that the microRNA expression profile was distinct from CD4(+) T-cell controls and B-cell lymphomas. The majority (104 of 114) of SzS-associated microRNAs (P < .05) were down-regulated and their expression pattern was largely consistent with previously reported genomic copy number abnormalities and were found to be highly enriched (P < .001) for aberrantly expressed target genes. Levels of miR-223 distinguished SzS samples (n = 32) from healthy controls (n = 19) and patients with mycosis fungoides (n = 11) in more than 90% of samples. Furthermore, we demonstrate that the down-regulation of intronically encoded miR-342 plays a role in the pathogenesis of SzS by inhibiting apoptosis, and describe a novel mechanism of regulation for this microRNA via binding of miR-199a* to its host gene. We also provide the first in vivo evidence for down-regulation of the miR-17-92 cluster in malignancy and demonstrate that ectopic miR-17-5p expression increases apoptosis and decreases cell proliferation in SzS cells.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Sezary Syndrome/genetics , Apoptosis , Blotting, Western , Cell Proliferation , Gene Expression Profiling , Humans , Luciferases/metabolism , Lymphoma, B-Cell/blood , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/genetics , MicroRNAs/genetics , Mycosis Fungoides/blood , Mycosis Fungoides/diagnosis , Mycosis Fungoides/genetics , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sezary Syndrome/blood , Sezary Syndrome/diagnosis , T-Lymphocytes/metabolism
15.
Br J Haematol ; 149(6): 844-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20408843

ABSTRACT

Refractory anaemia with ring sideroblasts (RARS) is characterized by anaemia, erythroid apoptosis, cytochrome c release and mitochondrial ferritin accumulation. Granulocyte-colony-stimulating factor (G-CSF) inhibits the first three of these features in vitro and in vivo. To dissect the molecular mechanisms underlying the RARS phenotype and anti-apoptotic effects of G-CSF, erythroblasts generated from normal (NBM) and RARS marrow CD34(+) cells were cultured +/-G-CSF and subjected to gene expression analysis (GEP). Several erythropoiesis-associated genes that were deregulated in RARS CD34(+) cells showed normal expression in erythroblasts, underscoring the importance of differentiation-specific GEP. RARS erythroblasts showed a marked deregulation of several pathways including apoptosis, DNA damage repair, mitochondrial function and the JAK/Stat pathway. ABCB7, transporting iron from mitochondria to cytosol and associated with inherited ring sideroblast formation was severely suppressed and expression decreased with differentiation, while increasing in NBM cultures. The same pattern was observed for the mitochondrial integrity gene MFN2. Other downregulated key genes included STAT5B, HSPA5, FANCC and the negative apoptosis regulator MAP3K7. Methylation status of key downregulated genes was normal. The mitochondrial pathway including MFN2 was significantly modified by G-CSF, and several heat shock protein genes were upregulated, as evidence of anti-apoptotic protection of erythropoiesis. By contrast, G-CSF had no effect on iron-transport or erythropoiesis-associated genes.


Subject(s)
Anemia, Refractory/metabolism , Anemia, Sideroblastic/metabolism , Erythroblasts/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Aged , Apoptosis/genetics , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Erythroblasts/drug effects , Erythroblasts/pathology , Erythroid Precursor Cells/pathology , Erythropoiesis/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Iron/metabolism , Male , Middle Aged , Mitochondria/physiology , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/genetics
17.
Nat Med ; 16(1): 59-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19966810

ABSTRACT

The identification of the genes associated with chromosomal translocation breakpoints has fundamentally changed understanding of the molecular basis of hematological malignancies. By contrast, the study of chromosomal deletions has been hampered by the large number of genes deleted and the complexity of their analysis. We report the generation of a mouse model for human 5q- syndrome using large-scale chromosomal engineering. Haploinsufficiency of the Cd74-Nid67 interval (containing Rps14, encoding the ribosomal protein S14) caused macrocytic anemia, prominent erythroid dysplasia and monolobulated megakaryocytes in the bone marrow. These effects were associated with defective bone marrow progenitor development, the appearance of bone marrow cells expressing high amounts of the tumor suppressor p53 and increased bone marrow cell apoptosis. Notably, intercrossing with p53-deficient mice completely rescued the progenitor cell defect, restoring common myeloid progenitor and megakaryocytic-erythroid progenitor, granulocyte-monocyte progenitor and hematopoietic stem cell bone marrow populations. This mouse model suggests that a p53-dependent mechanism underlies the pathophysiology of the 5q- syndrome.


Subject(s)
Anemia, Macrocytic/genetics , Chromosome Deletion , Disease Models, Animal , Genes, p53/genetics , Myelodysplastic Syndromes/genetics , Animals , Apoptosis/genetics , Chromosomes, Mammalian/genetics , Hematopoietic Stem Cells/physiology , Humans , Mice , Synteny/genetics
18.
Haematologica ; 94(12): 1762-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19797731

ABSTRACT

Clonal heterogeneity has not been described in patients with myelodysplastic syndrome with isolated del(5q), for which lenalidomide has emerged as a highly potent treatment. However, transformation to acute myeloid leukemia is occasionally observed, particularly in patients without a cytogenetic response to lenalidomide. We performed molecular studies in a patient with classical 5q- syndrome with complete erythroid and partial cytogenetic response to lenalidomide, who evolved to high-risk myelodysplastic syndrome with complex karyotype. Immunohistochemistry of pre-treatment marrow biopsies revealed a small fraction of progenitors with overexpression of p53 and sequencing confirmed a TP53 mutation. TP53 mutated subclones have not previously been described in myelodysplastic syndrome with isolated del(5q) and indicates a previously unknown heterogeneity of this disease. The aberrant subclone remained stable during the treatment with lenalidomide and expanded at transformation, suggesting that this pre-existing cell population had molecular features which made it insensitive to lenalidomide and prone to disease progression.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Myelodysplastic Syndromes/genetics , Aged , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Chromosome Banding , Clone Cells/metabolism , Clone Cells/pathology , Disease Progression , Female , Gene Expression Profiling , Genetic Heterogeneity , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Lenalidomide , Mutation , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/pathology , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Tumor Suppressor Protein p53/genetics
19.
Blood ; 114(17): 3538-45, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19692701

ABSTRACT

We studied patients with myeloid neoplasm associated with ringed sideroblasts and/or thrombocytosis. The combination of ringed sideroblasts 15% or greater and platelet count of 450 x 10(9)/L or greater was found in 19 subjects fulfilling the diagnostic criteria for refractory anemia with ringed sideroblasts (RARS) associated with marked thrombocytosis (RARS-T), and in 3 patients with primary myelofibrosis. JAK2 and MPL mutations were detected in circulating granulocytes and bone marrow CD34+ cells, but not in T lymphocytes, from 11 of 19 patients with RARS-T. Three patients with RARS, who initially had low to normal platelet counts, progressed to RARS-T, and 2 of them acquired JAK2 (V617F) at this time. In female patients with RARS-T, granulocytes carrying JAK2 (V617F) represented only a fraction of clonal granulocytes as determined by X-chromosome inactivation patterns. RARS and RARS-T patient groups both consistently showed up-regulation of ALAS2 and down-regulation of ABCB7 in CD34+ cells, but several other genes were differentially expressed, including PSIP1 (LEDGF), CXCR4, and CDC2L5. These observations suggest that RARS-T is indeed a myeloid neoplasm with both myelodysplastic and myeloproliferative features at the molecular and clinical levels and that it may develop from RARS through the acquisition of somatic mutations of JAK2, MPL, or other as-yet-unknown genes.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/pathology , Thrombocytosis/genetics , Thrombocytosis/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Female , Flow Cytometry , Gene Expression Profiling , Granulocytes/metabolism , Granulocytes/pathology , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Platelet Count , Receptors, Thrombopoietin/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , X Chromosome Inactivation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...