Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Blood Adv ; 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39374575

ABSTRACT

Adult haematopoietic stem cells (HSCs) are responsible for the lifelong production of blood and immune cells, a process regulated by extracellular cues including cytokines. Many cytokines signal through the conserved JAK/STAT pathway, in which tyrosine-phosphorylated STATs (pSTATs) function as transcription factors. STAT5 is a pivotal downstream mediator of several cytokines known to regulate haematopoiesis but its function in the HSC compartment remains poorly understood. Here, we show that STAT5-deficient HSCs exhibit an unusual phenotype: reduced multi-lineage repopulation and self-renewal, combined with reduced exit from quiescence and increased differentiation. This was driven not only by loss of canonical pSTAT5 signalling, but also by loss of distinct transcriptional functions mediated by STAT5 lacking canonical tyrosine phosphorylation (uSTAT5). Consistent with this concept, expression of an unphosphorylatable STAT5 mutant constrained wild-type HSC differentiation, promoted their maintenance and upregulated transcriptional programs associated with quiescence and stemness. The JAK1/2 inhibitor, ruxolitinib, which increased the uSTAT5:pSTAT5 ratio, had similar effects on murine HSC function: it constrained HSC differentiation and proliferation, promoted HSC maintenance and upregulated transcriptional programs associated with stemness. Ruxolitinib also enhanced serial replating of normal human HSPCs, CALR-mutant murine HSCs and HSPCs obtained from patients with myelofibrosis. Our results therefore reveal a previously unrecognized interplay between pSTAT5 and uSTAT5 in the control of HSC function and highlight JAK inhibition as a potential strategy for enhancing HSC function during ex vivo culture. Increased levels of uSTAT5 may also contribute to the failure of JAK inhibitors to eradicate myeloproliferative neoplasms.

2.
Exp Hematol ; 135: 104246, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763471

ABSTRACT

Key studies in pre-leukemic disorders have linked increases in pro-inflammatory cytokines with accelerated phases of the disease, but the precise role of the cellular microenvironment in disease initiation and evolution remains poorly understood. In myeloproliferative neoplasms (MPNs), higher levels of specific cytokines have been previously correlated with increased disease severity (tumor necrosis factor-alpha [TNF-α], interferon gamma-induced protein-10 [IP-10 or CXCL10]) and decreased survival (interleukin 8 [IL-8]). Whereas TNF-α and IL-8 have been studied by numerous groups, there is a relative paucity of studies on IP-10 (CXCL10). Here we explore the relationship of IP-10 levels with detailed genomic and clinical data and undertake a complementary cytokine screen alongside functional assays in a wide range of MPN mouse models. Similar to patients, levels of IP-10 were increased in mice with more severe disease phenotypes (e.g., JAK2V617F/V617F TET2-/- double-mutant mice) compared with those with less severe phenotypes (e.g., CALRdel52 or JAK2+/V617F mice) and wild-type (WT) littermate controls. Although exposure to IP-10 did not directly alter proliferation or survival in single hematopoietic stem cells (HSCs) in vitro, IP-10-/- mice transplanted with disease-initiating HSCs developed an MPN phenotype more slowly, suggesting that the effect of IP-10 loss was noncell-autonomous. To explore the broader effects of IP-10 loss, we crossed IP-10-/- mice into a series of MPN mouse models and showed that its loss reduces the erythrocytosis observed in mice with the most severe phenotype. Together, these data point to a potential role for blocking IP-10 activity in the management of MPNs.


Subject(s)
Chemokine CXCL10 , Myeloproliferative Disorders , Polycythemia , Animals , Humans , Male , Mice , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Disease Models, Animal , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice, Knockout , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/metabolism , Polycythemia/genetics , Polycythemia/pathology , Polycythemia/etiology , Female
3.
Nat Genet ; 56(2): 273-280, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38233595

ABSTRACT

Myeloproliferative neoplasms (MPNs) are chronic cancers characterized by overproduction of mature blood cells. Their causative somatic mutations, for example, JAK2V617F, are common in the population, yet only a minority of carriers develop MPN. Here we show that the inherited polygenic loci that underlie common hematological traits influence JAK2V617F clonal expansion. We identify polygenic risk scores (PGSs) for monocyte count and plateletcrit as new risk factors for JAK2V617F positivity. PGSs for several hematological traits influenced the risk of different MPN subtypes, with low PGSs for two platelet traits also showing protective effects in JAK2V617F carriers, making them two to three times less likely to have essential thrombocythemia than carriers with high PGSs. We observed that extreme hematological PGSs may contribute to an MPN diagnosis in the absence of somatic driver mutations. Our study showcases how polygenic backgrounds underlying common hematological traits influence both clonal selection on somatic mutations and the subsequent phenotype of cancer.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/diagnosis , Phenotype , Janus Kinase 2/genetics , Genetic Risk Score
4.
Nat Commun ; 14(1): 7725, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38001082

ABSTRACT

Current therapies for myeloproliferative neoplasms (MPNs) improve symptoms but have limited effect on tumor size. In preclinical studies, tamoxifen restored normal apoptosis in mutated hematopoietic stem/progenitor cells (HSPCs). TAMARIN Phase-II, multicenter, single-arm clinical trial assessed tamoxifen's safety and activity in patients with stable MPNs, no prior thrombotic events and mutated JAK2V617F, CALRins5 or CALRdel52 peripheral blood allele burden ≥20% (EudraCT 2015-005497-38). 38 patients were recruited over 112w and 32 completed 24w-treatment. The study's A'herns success criteria were met as the primary outcome ( ≥ 50% reduction in mutant allele burden at 24w) was observed in 3/38 patients. Secondary outcomes included ≥25% reduction at 24w (5/38), ≥50% reduction at 12w (0/38), thrombotic events (2/38), toxicities, hematological response, proportion of patients in each IWG-MRT response category and ELN response criteria. As exploratory outcomes, baseline analysis of HSPC transcriptome segregates responders and non-responders, suggesting a predictive signature. In responder HSPCs, longitudinal analysis shows high baseline expression of JAK-STAT signaling and oxidative phosphorylation genes, which are downregulated by tamoxifen. We further demonstrate in preclinical studies that in JAK2V617F+ cells, 4-hydroxytamoxifen inhibits mitochondrial complex-I, activates integrated stress response and decreases pathogenic JAK2-signaling. These results warrant further investigation of tamoxifen in MPN, with careful consideration of thrombotic risk.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Hematopoietic Stem Cells/metabolism , Signal Transduction , Neoplasms/metabolism , Tamoxifen/therapeutic use , Tamoxifen/metabolism , Mutation , Calreticulin/genetics , Calreticulin/metabolism
5.
Nat Commun ; 14(1): 5092, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37608017

ABSTRACT

Clonal tracking of cells using somatic mutations permits exploration of clonal dynamics in human disease. Here, we perform whole genome sequencing of 323 haematopoietic colonies from 10 individuals with the inherited ribosomopathy Shwachman-Diamond syndrome to reconstruct haematopoietic phylogenies. In ~30% of colonies, we identify mutually exclusive mutations in TP53, EIF6, RPL5, RPL22, PRPF8, plus chromosome 7 and 15 aberrations that increase SBDS and EFL1 gene dosage, respectively. Target gene mutations commence in utero, resulting in a profusion of clonal expansions, with only a few haematopoietic stem cell lineages (mean 8, range 1-24) contributing ~50% of haematopoietic colonies across 8 individuals (range 4-100% clonality) by young adulthood. Rapid clonal expansion during disease transformation is associated with biallelic TP53 mutations and increased mutation burden. Our study highlights how convergent somatic mutation of the p53-dependent nucleolar surveillance pathway offsets the deleterious effects of germline ribosomopathy but increases opportunity for TP53-mutated cancer evolution.


Subject(s)
Chromosomes, Human, Pair 7 , Germ Cells , Humans , Young Adult , Adult , Gene Dosage , Hematopoietic Stem Cells , Mutation
6.
Nat Cancer ; 4(8): 1193-1209, 2023 08.
Article in English | MEDLINE | ID: mdl-37550517

ABSTRACT

Aging facilitates the expansion of hematopoietic stem cells (HSCs) carrying clonal hematopoiesis-related somatic mutations and the development of myeloid malignancies, such as myeloproliferative neoplasms (MPNs). While cooperating mutations can cause transformation, it is unclear whether distinct bone marrow (BM) HSC-niches can influence the growth and therapy response of HSCs carrying the same oncogenic driver. Here we found different BM niches for HSCs in MPN subtypes. JAK-STAT signaling differentially regulates CDC42-dependent HSC polarity, niche interaction and mutant cell expansion. Asymmetric HSC distribution causes differential BM niche remodeling: sinusoidal dilation in polycythemia vera and endosteal niche expansion in essential thrombocythemia. MPN development accelerates in a prematurely aged BM microenvironment, suggesting that the specialized niche can modulate mutant cell expansion. Finally, dissimilar HSC-niche interactions underpin variable clinical response to JAK inhibitor. Therefore, HSC-niche interactions influence the expansion rate and therapy response of cells carrying the same clonal hematopoiesis oncogenic driver.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Aged , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Myeloproliferative Disorders/pathology , Bone Marrow/pathology , Bone Marrow/physiology , Hematopoietic Stem Cells/pathology , Bone and Bones/pathology , Tumor Microenvironment/genetics
7.
Blood ; 141(26): 3199-3214, 2023 06 29.
Article in English | MEDLINE | ID: mdl-36928379

ABSTRACT

Polycythemia vera (PV) is a myeloproliferative neoplasm driven by activating mutations in JAK2 that result in unrestrained erythrocyte production, increasing patients' hematocrit and hemoglobin concentrations, placing them at risk of life-threatening thrombotic events. Our genome-wide association study of 440 PV cases and 403 351 controls using UK Biobank data showed that single nucleotide polymorphisms in HFE known to cause hemochromatosis are highly associated with PV diagnosis, linking iron regulation to PV. Analysis of the FinnGen dataset independently confirmed overrepresentation of homozygous HFE variants in patients with PV. HFE influences the expression of hepcidin, the master regulator of systemic iron homeostasis. Through genetic dissection of mouse models of PV, we show that the PV erythroid phenotype is directly linked to hepcidin expression: endogenous hepcidin upregulation alleviates erythroid disease whereas hepcidin ablation worsens it. Furthermore, we demonstrate that in PV, hepcidin is not regulated by expanded erythropoiesis but is likely governed by inflammatory cytokines signaling via GP130-coupled receptors. These findings have important implications for understanding the pathophysiology of PV and offer new therapeutic strategies for this disease.


Subject(s)
Polycythemia Vera , Animals , Mice , Polycythemia Vera/genetics , Polycythemia Vera/complications , Hepcidins/genetics , Genome-Wide Association Study , Iron/metabolism , Phenotype , Homeostasis
8.
Nature ; 602(7895): 162-168, 2022 02.
Article in English | MEDLINE | ID: mdl-35058638

ABSTRACT

Mutations in cancer-associated genes drive tumour outgrowth, but our knowledge of the timing of driver mutations and subsequent clonal dynamics is limited1-3. Here, using whole-genome sequencing of 1,013 clonal haematopoietic colonies from 12 patients with myeloproliferative neoplasms, we identified 580,133 somatic mutations to reconstruct haematopoietic phylogenies and determine clonal histories. Driver mutations were estimated to occur early in life, including the in utero period. JAK2V617F was estimated to have been acquired by 33 weeks of gestation to 10.8 years of age in 5 patients in whom JAK2V617F was the first event. DNMT3A mutations were acquired by 8 weeks of gestation to 7.6 years of age in 4 patients, and a PPM1D mutation was acquired by 5.8 years of age. Additional genomic events occurred before or following JAK2V617F acquisition and as independent clonal expansions. Sequential driver mutation acquisition was separated by decades across life, often outcompeting ancestral clones. The mean latency between JAK2V617F acquisition and diagnosis was 30 years (range 11-54 years). Estimated historical rates of clonal expansion varied substantially (3% to 190% per year), increased with additional driver mutations, and predicted latency to diagnosis. Our study suggests that early driver mutation acquisition and life-long growth and evolution underlie adult myeloproliferative neoplasms, raising opportunities for earlier intervention and a new model for cancer development.


Subject(s)
Mutation , Myeloproliferative Disorders , Neoplasms , Adult , Child, Preschool , Clone Cells/pathology , Humans , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phylogeny , Protein Phosphatase 2C , Whole Genome Sequencing
9.
Haematologica ; 107(2): 489-499, 2022 02 01.
Article in English | MEDLINE | ID: mdl-33567811

ABSTRACT

Angioimmunoblastic T-cell lymphoma (AITL) and peripheral T-cell lymphoma with T follicular helper phenotype (PTCL-TFH) are a group of complex clinicopathological entities that originate from T follicular helper cells and share a similar mutation profile. Their diagnosis is often a challenge, particularly at an early stage, because of a lack of specific histological and immunophenotypic features, paucity of neoplastic T cells and prominent polymorphous infiltrate. We investigated whether the lymphoma-associated RHOA Gly17Val (c.50G>T) mutation, occurring in 60% of cases, is present in the early "reactive" lesions, and whether mutation analysis could help to advance the early diagnosis of lymphoma. The RHOA mutation was detected by quantitative polymerase chain reaction with a locked nucleic acid probe specific to the mutation, and a further peptide nucleic acid clamp oligonucleotide to suppress the amplification of the wild-type allele. The quantitative polymerase chain reaction assay was highly sensitive and specific, detecting RHOA Gly17Val at an allele frequency of 0.03%, but not other changes in Gly17, nor in 61 controls. Among the 37 cases of AITL and PTCL-TFH investigated, RHOA Gly17Val was detected in 62.2% (23/37) of which 19 had multiple biopsies including preceding biopsies in ten and follow-up biopsies in 11 cases. RHOA Gly17Val was present in each of these preceding or follow-up biopsies including 18 specimens that showed no evidence of lymphoma by combined histological, immunophenotypic and clonality analyses. The mutation was seen in biopsies 0-26.5 months (mean 7.87 months) prior to the lymphoma diagnosis. Our results show that RHOA Gly17Val mutation analysis is valuable in the early detection of AITL and PTCL-TFH.


Subject(s)
Immunoblastic Lymphadenopathy , Lymphoma, T-Cell, Peripheral , Lymphoma, T-Cell , Early Diagnosis , Humans , Immunoblastic Lymphadenopathy/diagnosis , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/pathology , Mutation , Phenotype , T-Lymphocytes, Helper-Inducer/pathology , rhoA GTP-Binding Protein/genetics
10.
Blood Adv ; 5(7): 1922-1932, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33821991

ABSTRACT

Calreticulin (CALR) is mutated in the majority of JAK2/MPL-unmutated myeloproliferative neoplasms (MPNs). Mutant CALR (CALRdel52) exerts its effect by binding to the thrombopoietin receptor MPL to cause constitutive activation of JAK-STAT signaling. In this study, we performed an extensive mutagenesis screen of the CALR globular N-domain and revealed 2 motifs critical for CALRdel52 oncogenic activity: (1) the glycan-binding lectin motif and (2) the zinc-binding domain. Further analysis demonstrated that the zinc-binding domain was essential for formation of CALRdel52 multimers, which was a co-requisite for MPL binding. CALRdel52 variants incapable of binding zinc were unable to homomultimerize, form CALRdel52-MPL heteromeric complexes, or stimulate JAK-STAT signaling. Finally, treatment with zinc chelation disrupted CALRdel52-MPL complexes in hematopoietic cells in conjunction with preferential eradication of cells expressing CALRdel52 relative to cells expressing other MPN oncogenes. In addition, zinc chelators exhibited a therapeutic effect in preferentially impairing growth of CALRdel52-mutant erythroblasts relative to unmutated erythroblasts in primary cultures of MPN patients. Together, our data implicate zinc as an essential cofactor for CALRdel52 oncogenic activity by enabling CALRdel52 multimerization and interaction with MPL, and suggests that perturbation of intracellular zinc levels may represent a new approach to abrogate the oncogenic activity of CALRdel52 in the treatment of MPNs.


Subject(s)
Calreticulin , Myeloproliferative Disorders , Calreticulin/genetics , Humans , Mutagenesis , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Receptors, Thrombopoietin/genetics , Zinc
11.
Hemasphere ; 4(3): e371, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32647796

ABSTRACT

Myeloproliferative neoplasms (MPNs) are characterized by deregulation of mature blood cell production and increased risk of myelofibrosis (MF) and leukemic transformation. Numerous driver mutations have been identified but substantial disease heterogeneity remains unexplained, implying the involvement of additional as yet unidentified factors. The inflammatory microenvironment has recently attracted attention as a crucial factor in MPN biology, in particular whether inflammatory cytokines and chemokines contribute to disease establishment or progression. Here we present a large-scale study of serum cytokine profiles in more than 400 MPN patients and identify an essential thrombocythemia (ET)-specific inflammatory cytokine signature consisting of Eotaxin, GRO-α, and EGF. Levels of 2 of these markers (GRO-α and EGF) in ET patients were associated with disease transformation in initial sample collection (GRO-α) or longitudinal sampling (EGF). In ET patients with extensive genomic profiling data (n = 183) cytokine levels added significant prognostic value for predicting transformation from ET to MF. Furthermore, CD56+CD14+ pro-inflammatory monocytes were identified as a novel source of increased GRO-α levels. These data implicate the immune cell microenvironment as a significant player in ET disease evolution and illustrate the utility of cytokines as potential biomarkers for reaching beyond genomic classification for disease stratification and monitoring.

12.
N Engl J Med ; 379(15): 1416-1430, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30304655

ABSTRACT

BACKGROUND: Myeloproliferative neoplasms, such as polycythemia vera, essential thrombocythemia, and myelofibrosis, are chronic hematologic cancers with varied progression rates. The genomic characterization of patients with myeloproliferative neoplasms offers the potential for personalized diagnosis, risk stratification, and treatment. METHODS: We sequenced coding exons from 69 myeloid cancer genes in patients with myeloproliferative neoplasms, comprehensively annotating driver mutations and copy-number changes. We developed a genomic classification for myeloproliferative neoplasms and multistage prognostic models for predicting outcomes in individual patients. Classification and prognostic models were validated in an external cohort. RESULTS: A total of 2035 patients were included in the analysis. A total of 33 genes had driver mutations in at least 5 patients, with mutations in JAK2, CALR, or MPL being the sole abnormality in 45% of the patients. The numbers of driver mutations increased with age and advanced disease. Driver mutations, germline polymorphisms, and demographic variables independently predicted whether patients received a diagnosis of essential thrombocythemia as compared with polycythemia vera or a diagnosis of chronic-phase disease as compared with myelofibrosis. We defined eight genomic subgroups that showed distinct clinical phenotypes, including blood counts, risk of leukemic transformation, and event-free survival. Integrating 63 clinical and genomic variables, we created prognostic models capable of generating personally tailored predictions of clinical outcomes in patients with chronic-phase myeloproliferative neoplasms and myelofibrosis. The predicted and observed outcomes correlated well in internal cross-validation of a training cohort and in an independent external cohort. Even within individual categories of existing prognostic schemas, our models substantially improved predictive accuracy. CONCLUSIONS: Comprehensive genomic characterization identified distinct genetic subgroups and provided a classification of myeloproliferative neoplasms on the basis of causal biologic mechanisms. Integration of genomic data with clinical variables enabled the personalized predictions of patients' outcomes and may support the treatment of patients with myeloproliferative neoplasms. (Funded by the Wellcome Trust and others.).


Subject(s)
Calreticulin/genetics , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Precision Medicine , Receptors, Thrombopoietin/genetics , Bayes Theorem , DNA, Neoplasm/analysis , Disease Progression , Disease-Free Survival , Humans , Multivariate Analysis , Myeloproliferative Disorders/classification , Phenotype , Prognosis , Proportional Hazards Models , Sequence Analysis, DNA
13.
Blood ; 127(4): 436-48, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26508782

ABSTRACT

The crucial dependence of chronic lymphocytic leukemia (CLL) cells on signals derived from the B cell receptor (BCR) has encouraged the development of new inhibitors, which interfere with BCR signaling and demonstrate clinical benefits in nearly all patients. In addition, signaling through Toll-like receptor (TLR) 9 of the innate immune system has been shown to further contribute to the activation of CLL cells. However, responses to TLR9 engagement are not uniform, but diametrically opposed with cell death in some patients and cell proliferation in others. We now provide evidence that heterogeneous responses to TLR agonists are related to differences in the ability of CLL cells to activate the BCR-associated kinase Syk. Notably, expression of ZAP-70 appears to be of crucial importance for TLR9-mediated activation of Syk. We show that the activation of Syk provides an antiapoptotic signal, which is independent of Mcl-1, Bcl-2, and Bcl-XL, but related to the degradation of the proapoptotic Bim. Mechanistically, TLR9-mediated antiapoptotic signals in ZAP-70-positive CLL trigger secretion of immunoglobulin M, which then serves as (auto-) antigen for a prosurvival BCR signal. Thus, our data show that single activation of the innate immune receptor TLR9 is sufficient to fully engage BCR signaling in ZAP-70-positive CLL, protecting malignant cells from apoptosis. We conclude that the integration of TLR signaling into an adaptive immune response can further promote survival of CLL cells and may contribute to the unfavorable prognosis of ZAP-70-positive CLL.


Subject(s)
Adaptive Immunity , Immunity, Innate , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , ZAP-70 Protein-Tyrosine Kinase/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line , Cell Survival , Humans , Intracellular Signaling Peptides and Proteins/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Protein-Tyrosine Kinases/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction , Syk Kinase , Toll-Like Receptor 9/immunology
14.
Nat Commun ; 6: 6691, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25849990

ABSTRACT

Clonal proliferation in myeloproliferative neoplasms (MPN) is driven by somatic mutations in JAK2, CALR or MPL, but the contribution of inherited factors is poorly characterized. Using a three-stage genome-wide association study of 3,437 MPN cases and 10,083 controls, we identify two SNPs with genome-wide significance in JAK2(V617F)-negative MPN: rs12339666 (JAK2; meta-analysis P=1.27 × 10(-10)) and rs2201862 (MECOM; meta-analysis P=1.96 × 10(-9)). Two additional SNPs, rs2736100 (TERT) and rs9376092 (HBS1L/MYB), achieve genome-wide significance when including JAK2(V617F)-positive cases. rs9376092 has a stronger effect in JAK2(V617F)-negative cases with CALR and/or MPL mutations (Breslow-Day P=4.5 × 10(-7)), whereas in JAK2(V617F)-positive cases rs9376092 associates with essential thrombocythemia (ET) rather than polycythemia vera (allelic χ(2) P=7.3 × 10(-7)). Reduced MYB expression, previously linked to development of an ET-like disease in model systems, associates with rs9376092 in normal myeloid cells. These findings demonstrate that multiple germline variants predispose to MPN and link constitutional differences in MYB expression to disease phenotype.


Subject(s)
Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Adult , Aged , Alleles , Calreticulin/genetics , Case-Control Studies , Cohort Studies , DNA-Binding Proteins/genetics , Female , GTP-Binding Proteins/genetics , Gene Frequency , Genes, myb/genetics , Genetic Predisposition to Disease , Genetic Variation , Genotype , HSP70 Heat-Shock Proteins/genetics , Humans , Janus Kinase 2/genetics , MDS1 and EVI1 Complex Locus Protein , Male , Middle Aged , Mutation , Myeloproliferative Disorders/genetics , Peptide Elongation Factors/genetics , Polymorphism, Single Nucleotide , Proto-Oncogenes/genetics , Receptors, Thrombopoietin/genetics , Telomerase/genetics , Transcription Factors/genetics
15.
N Engl J Med ; 372(7): 601-612, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25671252

ABSTRACT

BACKGROUND: Cancers result from the accumulation of somatic mutations, and their properties are thought to reflect the sum of these mutations. However, little is known about the effect of the order in which mutations are acquired. METHODS: We determined mutation order in patients with myeloproliferative neoplasms by genotyping hematopoietic colonies or by means of next-generation sequencing. Stem cells and progenitor cells were isolated to study the effect of mutation order on mature and immature hematopoietic cells. RESULTS: The age at which a patient presented with a myeloproliferative neoplasm, acquisition of JAK2 V617F homozygosity, and the balance of immature progenitors were all influenced by mutation order. As compared with patients in whom the TET2 mutation was acquired first (hereafter referred to as "TET2-first patients"), patients in whom the Janus kinase 2 (JAK2) mutation was acquired first ("JAK2-first patients") had a greater likelihood of presenting with polycythemia vera than with essential thrombocythemia, an increased risk of thrombosis, and an increased sensitivity of JAK2-mutant progenitors to ruxolitinib in vitro. Mutation order influenced the proliferative response to JAK2 V617F and the capacity of double-mutant hematopoietic cells and progenitor cells to generate colony-forming cells. Moreover, the hematopoietic stem-and-progenitor-cell compartment was dominated by TET2 single-mutant cells in TET2-first patients but by JAK2-TET2 double-mutant cells in JAK2-first patients. Prior mutation of TET2 altered the transcriptional consequences of JAK2 V617F in a cell-intrinsic manner and prevented JAK2 V617F from up-regulating genes associated with proliferation. CONCLUSIONS: The order in which JAK2 and TET2 mutations were acquired influenced clinical features, the response to targeted therapy, the biology of stem and progenitor cells, and clonal evolution in patients with myeloproliferative neoplasms. (Funded by Leukemia and Lymphoma Research and others.).


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/physiology , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins/genetics , Age of Onset , Cell Proliferation/genetics , DNA Mutational Analysis , Dioxygenases , Gene Expression , Homozygote , Humans , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Thrombosis/genetics , Transcription, Genetic , Up-Regulation
17.
J Clin Invest ; 123(5): 2169-82, 2013 May.
Article in English | MEDLINE | ID: mdl-23543057

ABSTRACT

Large regions of recurrent genomic loss are common in cancers; however, with a few well-characterized exceptions, how they contribute to tumor pathogenesis remains largely obscure. Here we identified primate-restricted imprinting of a gene cluster on chromosome 20 in the region commonly deleted in chronic myeloid malignancies. We showed that a single heterozygous 20q deletion consistently resulted in the complete loss of expression of the imprinted genes L3MBTL1 and SGK2, indicative of a pathogenetic role for loss of the active paternally inherited locus. Concomitant loss of both L3MBTL1 and SGK2 dysregulated erythropoiesis and megakaryopoiesis, 2 lineages commonly affected in chronic myeloid malignancies, with distinct consequences in each lineage. We demonstrated that L3MBTL1 and SGK2 collaborated in the transcriptional regulation of MYC by influencing different aspects of chromatin structure. L3MBTL1 is known to regulate nucleosomal compaction, and we here showed that SGK2 inactivated BRG1, a key ATP-dependent helicase within the SWI/SNF complex that regulates nucleosomal positioning. These results demonstrate a link between an imprinted gene cluster and malignancy, reveal a new pathogenetic mechanism associated with acquired regions of genomic loss, and underline the complex molecular and cellular consequences of "simple" cancer-associated chromosome deletions.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 20 , Gene Expression Regulation , Genomic Imprinting , Alleles , Animals , Cell Lineage , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Silencing , Heterozygote , Humans , Immediate-Early Proteins/genetics , Macaca , Macropodidae , Male , Models, Genetic , Multigene Family , Myeloproliferative Disorders/genetics , Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Repressor Proteins , Transcription, Genetic , Tumor Suppressor Proteins
18.
Methods Mol Biol ; 967: 115-31, 2013.
Article in English | MEDLINE | ID: mdl-23296725

ABSTRACT

Mutations in the JAK2 gene are prevalent in the human myeloid malignancies, being present in virtually all cases of polycythemia vera, and a significant proportion of patients with other myeloproliferative disorders. Various methods for the detection of acquired mutations in this gene are available depending on the need for sensitivity, quantification, or the ability to detect many different mutations. We summarize the various methods published and discuss their relative merits for each application. Two commonly used methods, quantitative real-time PCR (QPCR) for the detection of the JAK2 V617F mutation and high resolution melt-curve analysis (HRM) for the detection of multiple mutations within JAK2 exon 12, demonstrate the utility of each method and their limitations. The choice of methodology is dependent on the application; therefore there is no gold standard for detecting mutations in this gene.


Subject(s)
DNA Mutational Analysis/methods , Janus Kinase 2/genetics , Mutation , Base Sequence , Exons/genetics , Gold/chemistry , Humans , Nucleic Acid Denaturation , Oligonucleotide Probes/chemistry , Oligonucleotide Probes/genetics , Oligonucleotides/chemistry , Oligonucleotides/genetics , Quality Control , Reproducibility of Results , Transition Temperature
19.
Haematologica ; 91(8): 1100-4, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16885051

ABSTRACT

The acquired mutation Val617Phe in the tyrosine kinase JAK2 was recently identified in most but not all patients with classical myeloproliferative disorders. We describe a cytogenetic and molecular study of a JAK2Val617Phe-negative case of essential thrombocythemia harboring the acquired translocation t(X;5)(q13;q33). We show that this involves the inactive X-chromosome and is associated with silencing of autosomal genes within the adjacent 5q minus syndrome common deleted region. This is the first documented example of autosomal gene silencing adjacent to an X-autosome breakpoint in human malignancy and such a mechanism may underlie the pathogenesis of related disorders with translocations involving Xq13.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 5 , Chromosomes, Human, X , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Sex Chromosome Disorders/genetics , Thrombocythemia, Essential/genetics , Translocation, Genetic , Amino Acid Substitution , Base Sequence , Blotting, Southern , Bone Marrow/pathology , Breast Neoplasms/complications , Breast Neoplasms/genetics , Chromosome Disorders/complications , Chromosome Mapping , Chromosome Walking/methods , DNA Methylation , DNA Primers , Female , Humans , Janus Kinase 2 , Middle Aged , Phenylalanine , Reverse Transcriptase Polymerase Chain Reaction , Sex Chromosome Disorders/complications , Thrombocythemia, Essential/complications , Thrombocythemia, Essential/pathology , Valine
20.
Blood ; 108(10): 3548-55, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16873677

ABSTRACT

The identification of an acquired mutation of JAK2 in patients with myeloproliferative disorders has raised questions about the relationship between mutation-positive and mutation-negative subtypes, timing of the JAK2 mutation, and molecular mechanisms of disease progression. Here we demonstrate that patients with V617F(-) essential thrombocythemia do not commonly progress to become V617F(+). Consistent with the concept of distinct pathogenetic mechanisms, we show that patients with and without the JAK2 mutation have different patterns of cytogenetic abnormality, with virtually all patients carrying the 20q deletion or trisomy 9 being V617F(+). We also investigated the existence of a "pre-JAK2" phase by comparing the proportion of clonally derived granulocytes, estimated from X-chromosome inactivation patterns (XCIPs), with the proportion of V617F(+) granulocytes. Our results demonstrate that inherent XCIP variability between granulocytes and T cells produces a systematically biased pattern of results that may be misinterpreted as evidence for an excess of clonally derived granulocytes, an observation that limits the utility of XCIP analysis in this context. Lastly, we studied 4 patients with V617F(+) myeloproliferative disorders who subsequently developed acute myeloid leukemia. In 3 patients the leukemic cells were V617F(-), suggesting that in these patients the leukemia arose in a V617F(-) cell.


Subject(s)
Chromosome Aberrations , Janus Kinase 2/genetics , Leukemia/pathology , Mutation, Missense , Myeloproliferative Disorders/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Clone Cells/pathology , Disease Progression , Granulocytes/pathology , Humans , Leukemia/etiology , Leukemia/genetics , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/pathology , T-Lymphocytes/pathology , Time Factors , X Chromosome Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL