Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
Ann Hematol ; 102(12): 3383-3399, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37792065

ABSTRACT

Ruxolitinib (RUX) is a Janus kinase 1/2 inhibitor (JAKi) approved in the EU for treating disease­related splenomegaly or symptoms in adults patients with myelofibrosis (MF). This is an interim analysis of JAKoMo, a prospective, non­interventional, phase IV study in MF. Between 2012-2019 (cutoff March 2021), 928 patients (JAKi-naïve and -pretreated) enrolled from 122 German centers. This analysis focuses on JAKi-naïve patients. RUX was administered according to the Summary of Product Characteristics. Compared to the COMFORT-I, -II, and JUMP trials, patients in JAKoMo were older (median 73 years), had poorer Eastern Cooperative Oncology Group (ECOG) performance statuses (16.5% had ECOG ≥ 2), and were more transfusion dependent (48.5%). JAKoMo represents the more challenging patients with MF encountered outside of interventional studies. However, patients with low-risk International Prognostic Scoring System (IPSS) scores or without palpable splenomegaly were also included. Following RUX treatment, 82.5% of patients experienced rapid (≤ 1 month), significant decreases in palpable spleen size, which remained durable for 24 months (60% patients). Symptom assessment scores improved significantly in Month 1 (median -5.2) up to Month 12 (-6.2). Common adverse events (AEs) were anemia (31.2%) and thrombocytopenia (28.6%). At cutoff, 54.3% of patients had terminated the study due to, death, AEs, or deterioration of health. No new safety signals were observed. Interim analysis of the JAKoMo study confirms RUX safety and efficacy in a representative cohort of real-world, elderly, JAKi-naïve patients with MF. Risk scores were used in less than half of the patients to initiate RUX treatment.Trial registration: NCT05044026; September 14, 2021.


Subject(s)
Janus Kinase Inhibitors , Primary Myelofibrosis , Adult , Humans , Aged , Splenomegaly/drug therapy , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/drug therapy , Prospective Studies , Nitriles , Treatment Outcome
2.
Blood ; 131(18): 2065-2073, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29519804

ABSTRACT

The transcription factor "nuclear factor erythroid 2" (NFE2) is overexpressed in the majority of patients with myeloproliferative neoplasms (MPNs). In murine models, elevated NFE2 levels cause an MPN phenotype with spontaneous leukemic transformation. However, both the molecular mechanisms leading to NFE2 overexpression and its downstream targets remain incompletely understood. Here, we show that the histone demethylase JMJD1C constitutes a novel NFE2 target gene. JMJD1C levels are significantly elevated in polycythemia vera (PV) and primary myelofibrosis patients; concomitantly, global H3K9me1 and H3K9me2 levels are significantly decreased. JMJD1C binding to the NFE2 promoter is increased in PV patients, decreasing both H3K9me2 levels and binding of the repressive heterochromatin protein-1α (HP1α). Hence, JMJD1C and NFE2 participate in a novel autoregulatory loop. Depleting JMJD1C expression significantly reduced cytokine-independent growth of an MPN cell line. Independently, NFE2 is regulated through the epigenetic JAK2 pathway by phosphorylation of H3Y41. This likewise inhibits HP1α binding. Treatment with decitabine lowered H3Y41ph and augmented H3K9me2 levels at the NFE2 locus in HEL cells, thereby increasing HP1α binding, which normalized NFE2 expression selectively in JAK2V617F-positive cell lines.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Gene Expression , Myeloproliferative Disorders/genetics , NF-E2 Transcription Factor, p45 Subunit/genetics , Biomarkers , Chromobox Protein Homolog 5 , Cytokines/metabolism , DNA Methylation , Decitabine/pharmacology , Histones/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Models, Biological , Mutation , Myeloproliferative Disorders/metabolism , NF-E2 Transcription Factor, p45 Subunit/metabolism , Oxidoreductases, N-Demethylating/genetics , Phosphorylation , Polycythemia Vera/genetics , Promoter Regions, Genetic , Protein Binding
3.
Br J Haematol ; 182(6): 830-842, 2018 09.
Article in English | MEDLINE | ID: mdl-29974943

ABSTRACT

Knowledge of the molecular and clonal characteristics in the myelodysplastic syndromes (MDS) and during progression to acute myeloid leukaemia (AML) is essential to understand the disease dynamics and optimize treatment. Sequencing serial bone marrow samples of eight patients, we observed that MDS featured a median of 3 mutations. Mutations in genes involved in RNA-splicing or epigenetic regulation were most frequent, and exclusively present in the major clone. Minor subclones were distinguishable in three patients. As the MDS progressed, a median of one mutation was gained, leading to clonal outgrowth. No AML developed genetically independent of a pre-existing clone. The gained mutation mostly affected genes encoding signalling proteins. Additional acquisition of genomic aberrations frequently occurred. Upon treatment, emergence of new clones could be observed. As confirmed by single-cell sequencing, multiple mutations in identical genes in different clones were present within individual patients. DNA-methylation profiling in patients without identification of novel mutations in AML revealed methylation changes in individual genes. In conclusion, our data complement previous observations on the mutational and clonal characteristics in MDS and at progression. Moreover, DNA-methylation changes may be associated with progression in single patients. Redundancy of mutated genes in different clones suggests fertile grounds promoting clonal selection or acquisition.


Subject(s)
Clone Cells/pathology , Disease Progression , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Adult , DNA Methylation , Female , Humans , Leukemia, Myeloid, Acute/etiology , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/therapy , Single-Cell Analysis
4.
Blood ; 138(21): 2019-2020, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34821937

Subject(s)
Rosa , Farms , Mutation
5.
Histochem Cell Biol ; 147(1): 75-82, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27544663

ABSTRACT

Tissue microarray (TMA) technique is an established high-throughput method to analyze multiple tissue specimens in parallel. However, in order to obtain reliable results from immunohistochemical analyses of TMA blocks, cell composition of TMA spots must correspond to whole tissue sections (WTS) particularly in tissues with a heterogeneous cell composition as it is the case in myeloproliferative neoplasms (MPN). The aim of this study was to validate TMA of bone marrow biopsies from MPN patients. TMAs of MPN bone marrow biopsies (ET: n = 26, PV: n = 26, and PMF: n = 29) were compiled in triplicates and MPN-specific histological parameters were assessed. Results of TMA spots were compared with WTS' results using the intra-class correlation coefficient (ICC). Immunohistochemical NFE2 and calreticulin stainings of the TMA with quantitative evaluation were performed. TMA construction was technically successful with a loss of 10 % of all spots. ICC calculation revealed high to moderate correlations of TMA with WTS, especially the parameters that are typically affected in MPN tissue, e.g. cellularity of hematopoiesis (ICC 0.62-0.89), number of megakaryocytes (ICC 0.50-0.71), micro-vessel density (ICC 0.56-0.91), or grade of myelofibrosis (ICC 0.56-0.89). Results of NFE2 and calreticulin immunohistochemistry of MPN TMAs are consistent with previously published data. Overall, our results show moderate to good correlation between histological data of WTS and TMA spots illustrating that the TMA technique is applicable to bone marrow biopsies of MPN patients. However, TMA construction in triplicates is necessary to reach sufficient correlation. MPN TMAs can be applied for serial immunohistochemical surveys of archived tissues to assess the mutation status or to further sub-classify MPN cases.


Subject(s)
Bone Marrow/pathology , Myeloproliferative Disorders/pathology , Tissue Array Analysis , Adult , Aged , Aged, 80 and over , Biopsy , Humans , Middle Aged , Young Adult
6.
Haematologica ; 102(1): 85-93, 2017 01.
Article in English | MEDLINE | ID: mdl-27540137

ABSTRACT

The myeloproliferative neoplasms, including polycythemia vera, essential thrombocythemia and myelofibrosis, are distinguished by their debilitating symptom profiles, life-threatening complications and profound impact on quality of life. The role gender plays in the symptomatology of myeloproliferative neoplasms remains under-investigated. In this study we evaluated how gender relates to patients' characteristics, disease complications and overall symptom expression. A total of 2,006 patients (polycythemia vera=711, essential thrombocythemia=830, myelofibrosis=460, unknown=5) were prospectively evaluated, with patients completing the Myeloproliferative Neoplasm-Symptom Assessment Form and Brief Fatigue Inventory Patient Reported Outcome tools. Information on the individual patients' characteristics, disease complications and laboratory data was collected. Consistent with known literature, most female patients were more likely to have essential thrombocythemia (48.6% versus 33.0%; P<0.001) and most male patients were more likely to have polycythemia vera (41.8% versus 30.3%; P<0.001). The rate of thrombocytopenia was higher among males than females (13.9% versus 8.2%; P<0.001) and males also had greater red-blood cell transfusion requirements (7.3% versus 4.9%; P=0.02) with shorter mean disease duration (6.4 versus 7.2 years, P=0.03). Despite there being no statistical differences in risk scores, receipt of most therapies or prior complications (hemorrhage, thrombosis), females had more severe and more frequent symptoms for most individual symptoms, along with overall total symptom score (22.8 versus 20.3; P<0.001). Females had particularly high scores for abdominal-related symptoms (abdominal pain/discomfort) and microvascular symptoms (headache, fatigue, insomnia, concentration difficulties, dizziness; all P<0.01). Despite complaining of more severe symptom burden, females had similar quality of life scores to those of males. The results of this study suggest that gender contributes to the heterogeneity of myeloproliferative neoplasms by influencing phenotypic profiles and symptom expression.


Subject(s)
Myeloproliferative Disorders/epidemiology , Phenotype , Quality of Life , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/mortality , Prognosis , Sex Factors , Surveys and Questionnaires , Young Adult
7.
Blood ; 123(14): 2133-4, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24700715

ABSTRACT

In this issue of Blood, Lundberg et al correlate the presence of known mutations in patients with myeloproliferative neoplasms (MPNs) with clinical outcome, thereby proposing a molecular risk stratification.


Subject(s)
Clonal Evolution , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Female , Humans , Male
8.
Haematologica ; 101(9): 1054-64, 2016 09.
Article in English | MEDLINE | ID: mdl-27479815

ABSTRACT

We have recently demonstrated that the transcription factor nuclear factor-erythroid 2, which is critical for erythroid maturation and globin gene expression, plays an important role in the pathophysiology of myeloproliferative neoplasms. Myeloproliferative neoplasm patients display elevated levels of nuclear factor-erythroid 2 and transgenic mice overexpressing the transcription factor develop myeloproliferative neoplasm, albeit, surprisingly without erythrocytosis. Nuclear factor-erythroid 2 transgenic mice show both a reticulocytosis and a concomitant increase in iron deposits in the spleen, suggesting both enhanced erythrocyte production and increased red blood cell destruction. We therefore hypothesized that elevated nuclear factor-erythroid 2 levels may lead to increased erythrocyte destruction by interfering with organelle clearance during erythroid maturation. We have previously shown that nuclear factor-erythroid 2 overexpression delays erythroid maturation of human hematopoietic stem cells. Here we report that increased nuclear factor-erythroid 2 levels also impede murine maturation by retarding mitochondrial depolarization and delaying mitochondrial elimination. In addition, ribosome autophagy is delayed in transgenics. We demonstrate that the autophagy genes NIX and ULK1 are direct novel nuclear factor-erythroid 2 target genes, as these loci are bound by nuclear factor-erythroid 2 in chromatin immunoprecipitation assays. Moreover, Nix and Ulk1 expression is increased in transgenic mice and in granulocytes from polycythemia vera patients. This is the first report implying a role for nuclear factor-erythroid 2 in erythroid maturation by affecting autophagy.


Subject(s)
Autophagy , Erythroid Cells/cytology , Erythroid Cells/metabolism , Mitochondria/genetics , Mitochondria/metabolism , NF-E2 Transcription Factor/genetics , NF-E2 Transcription Factor/metabolism , Animals , Autophagy/genetics , Biomarkers , Erythropoiesis/drug effects , Erythropoiesis/genetics , Gene Expression , Gene Expression Regulation , Humans , Immunophenotyping , Membrane Potential, Mitochondrial , Mice , Mice, Transgenic , Phenylhydrazines/pharmacology , Polycythemia Vera/genetics , Polycythemia Vera/metabolism , Reticulocytes/cytology , Reticulocytes/drug effects , Reticulocytes/metabolism , Ribosomes/metabolism
9.
Blood ; 122(1): 93-9, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23670178

ABSTRACT

The World Health Organization (WHO) classification of myeloproliferative neoplasms (MPNs) comprises several entities including essential thrombocythemia (ET); primary myelofibrosis (PMF); and MPN, unclassifiable (MPN,U). Differential diagnosis between ET and early, prefibrotic PMF can be challenging but is critical because clinical course and outcome vary considerably between these entities. We have previously shown that the transcription factor nuclear factor erythroid 2 (NF-E2) is aberrantly expressed in MPN patients. Here we demonstrate that NF-E2 is mislocalized in PMF cells and that aberrant NF-E2 localization discriminates statistically highly significantly between ET and PMF. A threshold of 20% nuclear NF-E2 staining was cross-validated by ".682+ bootstrapping." Moreover, this cutoff correctly classifies diagnostic bone marrow biopsies of MPN,U patients specified upon follow-up as ET or PMF with 92% accuracy. Because interobserver concordance between independent pathologists was high (Spearman's rank correlation coefficient, 0.727), we propose that quantitative NF-E2 immunohistochemistry represents a diagnostic tool that can reliably support a differential diagnosis between ET and PMF.


Subject(s)
Erythroid Cells/metabolism , NF-E2 Transcription Factor, p45 Subunit/metabolism , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/pathology , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/pathology , Antigens, CD/metabolism , Biopsy , Cohort Studies , Diagnosis, Differential , Erythroid Cells/pathology , Humans , Immunohistochemistry/statistics & numerical data , Observer Variation , Prognosis , Receptors, Transferrin/metabolism , Tissue Banks
10.
Mediators Inflamm ; 2015: 101987, 2015.
Article in English | MEDLINE | ID: mdl-26543325

ABSTRACT

It has been known for some time that solid tumors, especially gastrointestinal tumors, can arise on the basis of chronic inflammation. However, the role of inflammation in the genesis of hematological malignancies has not been extensively studied. Recent evidence clearly shows that changes in the bone marrow niche can suffice to induce myeloid diseases. Nonetheless, while it has been demonstrated that myeloproliferative neoplasms (MPN) are associated with a proinflammatory state, it is not clear whether inflammatory processes contribute to the induction or maintenance of MPN. More provocatively stated: which comes first, the hen or the egg, inflammation or MPN? In other words, can chronic inflammation itself trigger an MPN? In this review, we will describe the evidence supporting a role for inflammation in initiating and promoting MPN development. Furthermore, we will compare and contrast the data obtained in gastrointestinal tumors with observations in MPN patients and models, pointing out the opportunities provided by novel murine MPN models to address fundamental questions regarding the role of inflammatory stimuli in the molecular pathogenesis of MPN.


Subject(s)
Cell Transformation, Neoplastic , Inflammation/diagnosis , Inflammation/pathology , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/pathology , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/pathology , Animals , Cell Proliferation , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Humans , Immune System , Mice
11.
Blood ; 119(5): 1096-7, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22308276

ABSTRACT

In this issue of Blood, Gautier and colleagues describe a novel signaling pathway in which deregulated JAK2 activity augments expression of a key regulator of the cell cycle, the CDC25A phosphatase, via a translational mechanism.

12.
Recent Results Cancer Res ; 201: 99-107, 2014.
Article in English | MEDLINE | ID: mdl-24756787

ABSTRACT

The establishment of imatinib as the standard therapy for CML marked the beginning of a new era of treatment. Due to occurring intolerance and resistance against the drug, developing newer inhibitors was promoted. This led to the second-generation inhibitors dasatinib, nilotinib and bosutinib. Despite all achieved improvement, all first- and second-generation inhibitors are ineffective against the BCR-ABL T315I "gatekeeper" mutation. In order to overcome this issue and to further improve the inhibitory effect, the third-generation inhibitor ponatinib was developed. Various clinical trials have been launched to study the effect of ponatinib in the clinical setting. Based on positive phase 1 and phase 2 trials, ponatinib was approved for the second-line treatment of CML and Ph+ ALL in December 2012 in the United States and in July 2013 in the European Union. Further trials investigate the potential effect of ponatinib in kinase-dependent subgroups of other malignancies. In conclusion, ponatinib has proved to be a powerful BCR-ABL inhibitor, which exhibits clinical activity both in BCR-ABL wild-type and mutant CML, including activity against the T315I mutation. Despite previous TKI failure, chronic-phase CML patients can achieve sustained remissions using the novel drug, offering a new therapeutic option in the treatment for CML.


Subject(s)
Antineoplastic Agents/therapeutic use , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Pyridazines/therapeutic use , Animals , Humans , Protein Kinase Inhibitors/therapeutic use
13.
Cancer Res ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885318

ABSTRACT

Increasing evidence supports the interplay between oncogenic mutations and immune escape mechanisms. Strategies to counteract the immune escape mediated by oncogenic signaling could provide improved therapeutic options for patients with various malignancies. As mutant calreticulin (CALR) is a common driver of myeloproliferative neoplasms (MPN), we analyzed the impact of oncogenic CALRdel52 on the bone marrow (BM) microenvironment in MPN. Single-cell RNA-sequencing revealed that CALRdel52 led to the expansion of TGF-ß1-producing erythroid progenitor cells and promoted the expansion of FoxP3+ regulatory T cells (Treg) in a murine MPN model. Treatment with an anti-TGF-ß antibody improved mouse survival and increased the glycolytic activity in CD4+ and CD8+ T cells in vivo, while T cell depletion abrogated the protective effects conferred by neutralizing TGF-ß. TGF-ß1 reduced perforin and TNF-α production by T cells in vitro. TGF-ß1 production by CALRdel52 cells was dependent on JAK1/2, PI3K, and ERK activity, which activated the transcription factor Sp1 to induce TGF-ß1 expression. In four independent patient cohorts, TGF-ß1 expression was increased in the BM of MPN patients compared to healthy individuals, and the BM of MPN patients contained a higher frequency of Treg compared to healthy individuals. Together, this study identified an ERK/Sp1/TGF-ß1 axis in CALRdel52 MPNs as a mechanism of immunosuppression that can be targeted to elicit T-cell-mediated cytotoxicity.

14.
J Clin Invest ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916965

ABSTRACT

Leukemia relapse is a major cause of death after allogeneic hematopoietic cell transplantation (allo-HCT). We tested the potential of targeting TIM-3 for improving graft-versus-leukemia (GVL) effects. We observed differential expression of TIM-3 ligands when hematopoietic stem cells overexpressed certain oncogenic-driver mutations. Anti-TIM-3 Ab-treatment improved survival of mice bearing leukemia with oncogene-induced TIM-3 ligand expression. Conversely, leukemia cells with low ligand expression were anti-TIM-3 treatment-resistant. In vitro, TIM-3 blockade or genetic deletion in CD8+ T cells (Tc) enhanced Tc activation, proliferation and IFN-γ production while enhancing GVL effects, preventing Tc exhaustion and improving Tc cytotoxicity and glycolysis in vivo. Conversely, TIM-3 deletion in myeloid cells did not affect allogeneic Tc proliferation and activation in vitro, suggesting that anti-TIM-3-treatment-mediated GVL effects are Tc-induced. In contrast to anti-PD-1 and anti-CTLA-4-treatment, anti-TIM-3-treatment did not enhance acute graft-versus-host-disease (aGVHD). TIM-3 and its ligands were frequently expressed in acute myeloid leukemia (AML) cells of patients with post-allo-HCT relapse. We deciphered the connection between oncogenic mutations found in AML and TIM-3 ligands expression and identify anti-TIM-3-treatment as a strategy to enhance GVL effects via metabolic and transcriptional Tc-reprogramming, without exacerbation of aGVHD. Our findings support clinical testing of anti-TIM-3 Abs in patients with AML relapse post-allo-HCT.

15.
Blood ; 118(24): 6392-8, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21860020

ABSTRACT

Proinflammatory cytokines such as TNFα are elevated in patients with myeloproliferative neoplasms (MPN), but their contribution to disease pathogenesis is unknown. Here we reveal a central role for TNFα in promoting clonal dominance of JAK2(V617F) expressing cells in MPN. We show that JAK2(V617F) kinase regulates TNFα expression in cell lines and primary MPN cells and TNFα expression is correlated with JAK2(V617F) allele burden. In clonogenic assays, normal controls show reduced colony formation in the presence of TNFα while colony formation by JAK2(V617F)-positive progenitor cells is resistant or stimulated by exposure to TNFα. Ectopic JAK2(V617F) expression confers TNFα resistance to normal murine progenitor cells and overcomes inherent TNFα hypersensitivity of Fanconi anemia complementation group C deficient progenitors. Lastly, absence of TNFα limits clonal expansion and attenuates disease in a murine model of JAK2(V617F)-positive MPN. Altogether our data are consistent with a model where JAK2(V617F) promotes clonal selection by conferring TNFα resistance to a preneoplastic TNFα sensitive cell, while simultaneously generating a TNFα-rich environment. Mutations that confer resistance to environmental stem cell stressors are a recognized mechanism of clonal selection and leukemogenesis in bone marrow failure syndromes and our data suggest that this mechanism is also critical to clonal selection in MPN.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Janus Kinase 2/metabolism , Myeloproliferative Disorders/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Substitution , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/blood , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Mutant Proteins/metabolism , Myeloid Progenitor Cells/metabolism , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Point Mutation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics
16.
Haematologica ; 98(7): 1073-80, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23445878

ABSTRACT

The transcription factor nuclear factor erythroid-2 is over-expressed in patients with myeloproliferative neoplasms irrespective of the presence of the JAK2(V617F) mutation. Our transgenic mouse model over-expressing nuclear factor erythroid-2, which recapitulates many features of myeloproliferative neoplasms including transformation to acute myeloid leukemia, clearly implicates this transcription factor in the pathophysiology of myeloproliferative neoplasms. Because the targets mediating nuclear factor erythroid-2 effects are not well characterized, we conducted microarray analysis of CD34(+) cells lentivirally transduced to over-express nuclear factor erythroid-2 or to silence this transcription factor via shRNA, in order to identify novel target genes. Here, we report that the cytokine interleukin 8 is a novel target gene. Nuclear factor erythroid-2 directly binds the interleukin 8 promoter in vivo, and these binding sites are required for promoter activity. Serum levels of interleukin 8 are known to be elevated in both polycythemia vera and primary myelofibrosis patients. Recently, increased interleukin 8 levels have been shown to be predictive of inferior survival in primary myelofibrosis patients in multivariate analysis. Therefore, one of the mechanisms by which nuclear factor erythroid-2 contributes to myeloproliferative neoplasm pathology may be increased interleukin 8 expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-8/biosynthesis , Myelodysplastic-Myeloproliferative Diseases/metabolism , NF-E2 Transcription Factor, p45 Subunit/physiology , Animals , Antigens, CD34/genetics , Gene Targeting/methods , Genetic Vectors/administration & dosage , Humans , Interleukin-8/genetics , Lentivirus/genetics , Mice , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/genetics , Predictive Value of Tests , Protein Binding/genetics , Treatment Outcome , Tumor Cells, Cultured , U937 Cells
17.
Blood Adv ; 7(24): 7525-7538, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37639313

ABSTRACT

Leukemia stem cells (LSCs) share numerous features with healthy hematopoietic stem cells (HSCs). G-protein coupled receptor family C group 5 member C (GPRC5C) is a regulator of HSC dormancy. However, GPRC5C functionality in acute myeloid leukemia (AML) is yet to be determined. Within patient AML cohorts, high GPRC5C levels correlated with poorer survival. Ectopic Gprc5c expression increased AML aggression through the activation of NF-κB, which resulted in an altered metabolic state with increased levels of intracellular branched-chain amino acids (BCAAs). This onco-metabolic profile was reversed upon loss of Gprc5c, which also abrogated the leukemia-initiating potential. Targeting the BCAA transporter SLC7A5 with JPH203 inhibited oxidative phosphorylation and elicited strong antileukemia effects, specifically in mouse and patient AML samples while sparing healthy bone marrow cells. This antileukemia effect was strengthened in the presence of venetoclax and azacitidine. Our results indicate that the GPRC5C-NF-κB-SLC7A5-BCAAs axis is a therapeutic target that can compromise leukemia stem cell function in AML.


Subject(s)
Amino Acids, Branched-Chain , Leukemia, Myeloid, Acute , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Amino Acids, Branched-Chain/therapeutic use , Large Neutral Amino Acid-Transporter 1/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , NF-kappa B/metabolism , Receptors, G-Protein-Coupled/metabolism
18.
Blood ; 116(2): 254-66, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20339092

ABSTRACT

The transcription factor NF-E2 is overexpressed in the majority of patients with polycythemia vera (PV). Concomitantly, 95% of these patients carry the JAK2(V617F) mutation. Although NF-E2 levels correlate with JAK2(V671F) allele burden in some PV cohorts, the molecular mechanism causing aberrant NF-E2 expression has not been described. Here we show that NF-E2 expression is also increased in patients with essential thrombocythemia and primary myelofibrosis independent of the presence of the JAK2(V617F) mutation. Characterization of the NF-E2 promoter revealed multiple functional binding sites for AML1/RUNX-1. Chromatin immunoprecipitation demonstrated AML1 binding to the NF-E2 promoter in vivo. Moreover, AML1 binding to the NF-E2 promoter was significantly increased in granulocytes from PV patients compared with healthy controls. AML1 mRNA expression was elevated in patients with PV, essential thrombocythemia, and primary myelofibrosis both in the presence and absence of JAK2(V617F). In addition, AML1 and NF-E2 expression were highly correlated. RNAi-mediated suppression of either AML1 or of its binding partner CBF-beta significantly decreased NF-E2 expression. Moreover, expression of the leukemic fusion protein AML/ETO drastically decreased NF-E2 protein levels. Our data identify NF-E2 as a novel AML1 target gene and delineate a role for aberrant AML1 expression in mediating elevated NF-E2 expression in MPN patients.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Neoplastic , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , NF-E2 Transcription Factor, p45 Subunit/genetics , Blotting, Northern , Blotting, Western , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/biosynthesis , Electrophoretic Mobility Shift Assay , Gene Expression , Humans , Janus Kinase 2/metabolism , Myeloproliferative Disorders/metabolism , NF-E2 Transcription Factor, p45 Subunit/biosynthesis , Promoter Regions, Genetic , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Br J Haematol ; 153(5): 612-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21488857

ABSTRACT

The leukaemia-specific fusion oncoprotein RUNX1/RUNX1T1 (AML1/ETO), resulting from the chromosomal translocation (8;21) in acute myeloid leukaemia (AML), imposes a striking genotype-phenotype relationship upon this distinct subtype of AML, which is mediated by multiple, co-ordinate downstream effects induced by this chimeric transcription factor. We previously identified the LAT2 gene, encoding the adaptor molecule LAT2 (NTAL, LAB), which is phosphorylated by KIT and has a role in mast cell and B-cell activation, as a target of the repressor activity of RUNX1/RUNX1T1. These results were confirmed and extended by demonstrating downregulation of the LAT2 protein in response to conditional RUNX1/RUNX1T1 expression, and its absence in primary AML with the t(8;21). In contrast, in a cohort of 43 AML patients, higher levels of LAT2 were associated with myelomonocytic features. Differentiation of HL-60 and NB4 cells towards granulocytes by all trans-retinoic acid (ATRA) resulted in downregulation of LAT2; conversely, it was upregulated during phorbol ester-induced monocytic differentiation of HL-60 cells. Forced expression of LAT2 in Kasumi-1 cells resulted in a striking block of ATRA- and phorbol ester-induced differentiation, implicating disturbances of the graded expression of this adaptor molecule in the maturation block of myeloid leukaemia cells.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Core Binding Factor Alpha 2 Subunit/physiology , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/physiology , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Cell Differentiation/drug effects , Cholecalciferol/pharmacology , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic , Granulocytes/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phorbol Esters/pharmacology , RUNX1 Translocation Partner 1 Protein , Translocation, Genetic , Tumor Cells, Cultured , Up-Regulation/drug effects
20.
Proc Natl Acad Sci U S A ; 105(13): 5177-82, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18362329

ABSTRACT

The giant cytosolic protease tripeptidyl peptidase II (TPPII) has been implicated in the regulation of proliferation and survival of malignant cells, particularly lymphoma cells. To address its functions in normal cellular and systemic physiology we have generated TPPII-deficient mice. TPPII deficiency activates cell type-specific death programs, including proliferative apoptosis in several T lineage subsets and premature cellular senescence in fibroblasts and CD8(+) T cells. This coincides with up-regulation of p53 and dysregulation of NF-kappaB. Prominent degenerative alterations at the organismic level were a decreased lifespan and symptoms characteristic of immunohematopoietic senescence. These symptoms include accelerated thymic involution, lymphopenia, impaired proliferative T cell responses, extramedullary hematopoiesis, and inflammation. Thus, TPPII is important for maintaining normal cellular and systemic physiology, which may be relevant for potential therapeutic applications of TPPII inhibitors.


Subject(s)
Aging/immunology , Apoptosis/immunology , Serine Endopeptidases/deficiency , Serine Endopeptidases/metabolism , Aminopeptidases , Animals , Cell Differentiation/immunology , Cells, Cultured , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Fibroblasts , Gene Deletion , Lymphopenia/enzymology , Lymphopenia/genetics , Lymphopenia/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Phenotype , Serine Endopeptidases/genetics , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/enzymology , Thymus Gland/immunology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL