Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Sci Rep ; 12(1): 18656, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333382

ABSTRACT

Advanced computational methods exploit gene expression and epigenetic datasets to predict gene regulatory networks controlled by transcription factors (TFs). These methods have identified cell fate determining TFs but require large amounts of reference data and experimental expertise. Here, we present an easy to use network-based computational framework that exploits enhancers defined by bidirectional transcription, using as sole input CAGE sequencing data to correctly predict TFs key to various human cell types. Next, we applied this Analysis Algorithm for Networks Specified by Enhancers based on CAGE (ANANSE-CAGE) to predict TFs driving red and white blood cell development, and THP-1 leukemia cell immortalization. Further, we predicted TFs that are differentially important to either cell line- or primary- associated MLL-AF9-driven gene programs, and in primary MLL-AF9 acute leukemia. Our approach identified experimentally validated as well as thus far unexplored TFs in these processes. ANANSE-CAGE will be useful to identify transcription factors that are key to any cell fate change using only CAGE-seq data as input.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Algorithms , Blood Cells/metabolism , Computational Biology
2.
Ann Hematol ; 101(3): 571-579, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35024892

ABSTRACT

Acute myeloid leukemia (AML) is a highly heterogeneous disease showing dynamic clonal evolution patterns over time. Various subclones may be present simultaneously and subclones may show a different expansion pattern and respond differently to applied therapies. It is already clear that immunophenotyping and genetic analyses may yield overlapping, but also complementary information. Detailed information on the genetic make-up of immunophenotypically defined subclones is however scarce. We performed error-corrected sequencing for 27 myeloid leukemia driver genes in 86, FACS-sorted immunophenotypically characterized normal and aberrant subfractions in 10 AML patients. We identified three main scenarios. In the first group of patients, the two techniques were equally well characterizing the malignancy. In the second group, most of the isolated populations did not express aberrant immunophenotypes but still harbored several genetic aberrancies, indicating that the information obtained only by immunophenotyping would be incomplete. Vice versa, one patient was identified in which genetic mutations were found only in a small fraction of the immunophenotypically defined malignant populations, indicating that the genetic analysis gave an incomplete picture of the disease. We conclude that currently, characterization of leukemic cells in AML by molecular and immunophenotypic techniques is complementary, and infer that both techniques should be used in parallel in order to obtain the most complete view on the disease.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Clonal Evolution , Gene Expression Regulation, Leukemic , Genetic Variation , Humans , Immunophenotyping , Mutation
3.
Blood ; 138(1): 86-90, 2021 07 08.
Article in English | MEDLINE | ID: mdl-33690840

ABSTRACT

Mutations in the transcription factors GATA binding factor 1 (GATA1), growth factor independence 1B (GFI1B), and Runt-related transcription factor 1 (RUNX1) cause familial platelet and bleeding disorders. Mutant platelets exhibit common abnormalities including an α-granule reduction resulting in a grayish appearance in blood smears. This suggests that similar pathways are deregulated by different transcription factor mutations. To identify common factors, full platelet proteomes from 11 individuals with mutant GATA1R216Q, GFI1BQ287*, RUNX1Q154Rfs, or RUNX1TD2-6 and 28 healthy controls were examined by label-free quantitative mass spectrometry. In total, 2875 platelet proteins were reliably quantified. Clustering analysis of more than 300 differentially expressed proteins revealed profound differences between cases and controls. Among cases, 44 of 143 significantly downregulated proteins were assigned to platelet function, hemostasis, and granule biology, in line with platelet dysfunction and bleedings. Remarkably, none of these proteins were significantly diminished in all affected cases. Similarly, no proteins were commonly overrepresented in all affected cases compared with controls. These data indicate that the studied transcription factor mutations alter platelet proteomes in distinct largely nonoverlapping manners. This work provides the quantitative landscape of proteins that affect platelet function when deregulated by mutated transcription factors in inherited bleeding disorders.


Subject(s)
Blood Platelet Disorders/metabolism , Blood Platelets/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , GATA1 Transcription Factor/metabolism , Proteome/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Homeostasis , Humans , Mutation/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Leukemia ; 32(3): 828-836, 2018 03.
Article in English | MEDLINE | ID: mdl-28871137

ABSTRACT

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Subject(s)
Gene Rearrangement , Leukemia, Myeloid, Acute/genetics , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Transcriptional Activation , Translocation, Genetic , Cell Line , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , Epigenesis, Genetic , Exons , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Introns , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic
5.
Oncogene ; 36(23): 3346-3356, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28114278

ABSTRACT

In 11q23 leukemias, the N-terminal part of the mixed lineage leukemia (MLL) gene is fused to >60 different partner genes. In order to define a core set of MLL rearranged targets, we investigated the genome-wide binding of the MLL-AF9 and MLL-AF4 fusion proteins and associated epigenetic signatures in acute myeloid leukemia (AML) cell lines THP-1 and MV4-11. We uncovered both common as well as specific MLL-AF9 and MLL-AF4 target genes, which were all marked by H3K79me2, H3K27ac and H3K4me3. Apart from promoter binding, we also identified MLL-AF9 and MLL-AF4 binding at specific subsets of non-overlapping active distal regulatory elements. Despite this differential enhancer binding, MLL-AF9 and MLL-AF4 still direct a common gene program, which represents part of the RUNX1 gene program and constitutes of CD34+ and monocyte-specific genes. Comparing these data sets identified several zinc finger transcription factors (TFs) as potential MLL-AF9 co-regulators. Together, these results suggest that MLL fusions collaborate with specific subsets of TFs to deregulate the RUNX1 gene program in 11q23 AMLs.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Adult , Core Binding Factor Alpha 2 Subunit/genetics , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Prognosis , Promoter Regions, Genetic
6.
Leukemia ; 31(1): 26-33, 2017 01.
Article in English | MEDLINE | ID: mdl-27416910

ABSTRACT

Post-remission treatment (PRT) in patients with cytogenetically normal (CN) acute myeloid leukemia (AML) in first complete remission (CR1) is debated. We studied 521 patients with CN-AML in CR1, for whom mutational status of NPM1 and FLT3-ITD was available, including the FLT3-ITD allelic ratio. PRT consisted of reduced intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation (alloHSCT) (n=68), myeloablative conditioning (MAC) alloHSCT (n=137), autologous hematopoietic stem cell transplantation (autoHSCT) (n=168) or chemotherapy (n=148). Favorable overall survival (OS) was found for patients with mutated NPM1 without FLT3-ITD (71±4%). Outcome in patients with a high FLT3-ITD allelic ratio appeared to be very poor with OS and relapse-free survival (RFS) of 23±8% and 12±6%, respectively. Patients with wild-type NPM1 without FLT3-ITD or with a low allelic burden of FLT3-ITD were considered as intermediate-risk group because of similar OS and RFS at 5 years, in which PRT by RIC alloHSCT resulted in better OS and RFS as compared with chemotherapy (hazard ratio (HR) 0.56, P=0.022 and HR 0.50, P=0.004, respectively) or autoHSCT (HR 0.60, P=0.046 and HR 0.60, P=0.043, respectively). The lowest cumulative incidence of relapse (23±4%) was observed following MAC alloHSCT. These results suggest that alloHSCT may be preferred in patients with molecularly intermediate-risk CN-AML, while the choice of conditioning type may be personalized according to risk for non-relapse mortality.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Adult , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mutation , Nucleophosmin , Precision Medicine/methods , Remission Induction , Risk Assessment , Survival Rate , Tandem Repeat Sequences , Transplantation Conditioning/methods , Young Adult
8.
Leukemia ; 27(10): 2032-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23860450

ABSTRACT

Reliable detection of JAK2-V617F is critical for accurate diagnosis of myeloproliferative neoplasms (MPNs); in addition, sensitive mutation-specific assays can be applied to monitor disease response. However, there has been no consistent approach to JAK2-V617F detection, with assays varying markedly in performance, affecting clinical utility. Therefore, we established a network of 12 laboratories from seven countries to systematically evaluate nine different DNA-based quantitative PCR (qPCR) assays, including those in widespread clinical use. Seven quality control rounds involving over 21,500 qPCR reactions were undertaken using centrally distributed cell line dilutions and plasmid controls. The two best-performing assays were tested on normal blood samples (n=100) to evaluate assay specificity, followed by analysis of serial samples from 28 patients transplanted for JAK2-V617F-positive disease. The most sensitive assay, which performed consistently across a range of qPCR platforms, predicted outcome following transplant, with the mutant allele detected a median of 22 weeks (range 6-85 weeks) before relapse. Four of seven patients achieved molecular remission following donor lymphocyte infusion, indicative of a graft vs MPN effect. This study has established a robust, reliable assay for sensitive JAK2-V617F detection, suitable for assessing response in clinical trials, predicting outcome and guiding management of patients undergoing allogeneic transplant.


Subject(s)
Janus Kinase 2/genetics , Mutation/genetics , Myeloproliferative Disorders/genetics , Neoplasm Recurrence, Local/diagnosis , Neoplasm, Residual/diagnosis , Real-Time Polymerase Chain Reaction , Adult , Aged , Cytogenetic Analysis , Europe , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myeloproliferative Disorders/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm, Residual/genetics , Prognosis , RNA, Messenger/genetics , Remission Induction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation , Transplantation, Homologous , Young Adult
9.
Bone Marrow Transplant ; 46(8): 1071-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21042305

ABSTRACT

Five patients with adult-onset metachromatic leukodystrophy (MLD) underwent allo-SCT. Conditioning was reduced in intensity and grafts were obtained from voluntary unrelated donors. All but one graft were depleted of T-lymphocytes. Patient age at transplantation varied from 18 to 29 (median, 27) years. Two patients rejected their graft and MLD progressed. The recipient of the unmanipulated graft converted to complete donor chimerism with normalization of arylsulphatase A (ARSA) levels. Despite ARSA normalization, he deteriorated. Another patient was a mixed chimera. Following escalated doses of donor lymphocyte infusions he converted to complete donor chimerism. His levels of ARSA correlated positively with the percentage of donor cells and MLD was not progressive. The fifth patient died after 35 days from complications associated with GVHD. We conclude that results of allo-SCT in symptomatic MLD patients are poor. However, allo-SCT may stop progression of MLD in selected patients.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukodystrophy, Metachromatic/surgery , Adolescent , Adult , Female , Humans , Male , Young Adult
10.
Leukemia ; 24(11): 1834-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861919

ABSTRACT

Transcription factor Growth factor independence 1 (Gfi1) is required for multilineage blood cell development, from stem and progenitor cells to differentiated lymphoid and myeloid cells. Gfi1 expression is rapidly induced by cytokines that control both the adaptive and innate immune systems. Gfi1 itself represses the expression of genes implicated in cell survival, proliferation and differentiation. Changes in Gfi1 expression and function have not only been implicated in neutropenia, allergy, autoimmunity and hyperinflammatory responses, but also in lymphoma and more recently in the development of leukemia. In this study, we review how Gfi1 and its paralogue Gfi1b control the development of blood cells, discuss how changes in Gfi1 and Gfi1b function contribute to hematological disease and report on the molecular function of these proteins.


Subject(s)
DNA-Binding Proteins/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Cell Differentiation , Cytokines/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Humans , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Zinc Fingers/physiology
12.
Leukemia ; 23(8): 1480-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19340006

ABSTRACT

Ubiquitination plays a major role in many aspects of hematopoiesis. Alterations in ubiquitination have been implicated in hematological cancer. The ubiquitin ligase Triad1 controls the proliferation of myeloid cells. Here, we show that two RING (really interesting new gene) domains in Triad1 differentially bind ubiquitin-conjugating enzymes, UbcH7 and Ubc13. UbcH7 and Ubc13 are known to catalyze the formation of different poly-ubiquitin chains. These chains mark proteins for proteasomal degradation or serve crucial non-proteolytic functions, respectively. In line with the dual Ubc interactions, we observed that Triad1 catalyzes the formation of both types of ubiquitin chains. The biological relevance of this finding was studied by testing Triad1 mutants in myeloid clonogenic assays. Full-length Triad1 and three mutants lacking conserved domains inhibited myeloid colony formation by over 50%. Strikingly, deletion of either RING finger completely abrogated the inhibitory effect of Triad1 in clonogenic growth. We conclude that Triad1 exhibits dual ubiquitin ligase activity and that both of its RING domains are crucial to inhibit myeloid cell proliferation. The differential interaction of the RINGs with Ubcs strongly suggests that the ubiquitination mediated through UbcH7 as well as Ubc13 plays a major role in myelopoiesis.


Subject(s)
Myelopoiesis/physiology , Protein Interaction Mapping , RING Finger Domains , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Kidney , Mice , NIH 3T3 Cells , Protein Binding , Protein Processing, Post-Translational , Recombinant Fusion Proteins/physiology , Structure-Activity Relationship , Two-Hybrid System Techniques , U937 Cells/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
Bone Marrow Transplant ; 44(4): 243-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19219079

ABSTRACT

Single nucleotide polymorphisms (SNPs) in the NOD2 gene have significant impact on both treatment-related mortality (TRM) and acute GVHD (aGVHD) in haematopoietic stem cell transplantation (HSCT). The effect of these polymorphisms when using T-cell-depleted grafts has been poorly studied. We retrospectively analysed NOD2 polymorphisms in a cohort of 85 patients and donors who received an HLA-identical sibling partially T-cell-depleted HSCT (0.5 x 10(6) CD3+ T cells per kg) following idarubicin-containing conditioning regimens. NOD2 polymorphisms were present in 14 of 85 (16.5%) of patients and 18 of 85 (21%) of donors. The risk of severe aGVHD (grade III-IV) and the 1-year TRM was significantly higher in the presence of NOD2 polymorphisms (hazard ratio (HR) 6.0, P=0.02 for severe aGVHD and HR 3.3, P=0.02 for TRM, respectively) and was most prominent in cases where patient and donor both had a polymorphism (HR 10.5, P=0.002 and HR 3.9, P=0.002). There was also a trend towards increased risk of bacteraemia due to coagulase-negative staphylococci in patients with an NOD2 polymorphism. We conclude that NOD2 polymorphism screening should be used to optimize donor selection and antimicrobial prophylaxis to reduce the occurrence of aGVHD and TRM following allogeneic HSCT.


Subject(s)
Graft vs Host Disease/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Nod2 Signaling Adaptor Protein/genetics , Adult , Cohort Studies , Gene Frequency , Genetic Variation , Graft vs Host Disease/immunology , Humans , Immunity, Innate , Lymphocyte Depletion , Male , Middle Aged , Nod2 Signaling Adaptor Protein/immunology , Polymorphism, Single Nucleotide , Retrospective Studies , Tissue Donors , Young Adult
17.
Leukemia ; 20(9): 1511-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16926849

ABSTRACT

The modification of proteins with ubiquitin is involved in the regulation of various important biological pathways. A crucial step in this process is the modification of specific substrate proteins with ubiquitin by E3 ligases. The ubiquitylation of proteins can result in altered protein function or degradation by the 26S proteasome. Various proteins playing an important role during hematopoiesis are regulated via ubiquitin modification. Recently, alterations in ubiquitylation and proteasomal degradation have been implicated in hematological cancers. Based on these findings, novel therapies that specifically target ubiquitylation or the proteasome are currently being developed. In this review, we will highlight the role of ubiquitylation in normal and malignant hematopoiesis and discuss novel therapeutical approaches that are now tested in various hematological malignancies.


Subject(s)
Hematopoiesis , Ubiquitin/metabolism , Humans , Hydrolysis , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction
18.
Ned Tijdschr Geneeskd ; 150(21): 1188-92, 2006 May 27.
Article in Dutch | MEDLINE | ID: mdl-16768284

ABSTRACT

For the last 2 years, a 55-year-old man had painful, recurrent oral ulcers. Histological examination showed non-specific inflammation. Eosinophilia in the blood and bone marrow raised the suspicion of hypereosinophilic syndrome. No other specific organ involvement was observed. The diagnosis was confirmed by detection of the fusion gene 'FIP1-like-1-platelet-derived growth factor receptor alpha' (FIP1L1-PDGFRA) in the peripheral blood and bone marrow. Treatment with the tyrosine-kinase inhibitor imatinib resulted in a rapid response that has been maintained for more than 2 years. Hypereosinophilic syndrome is a rare haematological disorder. Until recently diagnosis was made by exclusion, and the course of disease was often fatal. Fusion of the FIP1L1 gene to the PDGFRA gene was identified recently in some patients with hypereosinophilic syndrome. The fusion results in a novel tyrosine kinase that is constitutively activated and may induce proliferation ofhaematopoietic cells. Treatment with imatinib targets this tyrosine kinase. These advances in our understanding of the molecular biology of the disease will lead to a new classification of hypereosinophilic syndrome with specific therapeutic options.


Subject(s)
Hypereosinophilic Syndrome/drug therapy , Oral Ulcer/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Benzamides , Humans , Hypereosinophilic Syndrome/complications , Hypereosinophilic Syndrome/genetics , Imatinib Mesylate , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Oral Ulcer/etiology , Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
20.
Leukemia ; 19(5): 799-805, 2005 May.
Article in English | MEDLINE | ID: mdl-15744343

ABSTRACT

Acute promyelocytic leukemia (APL) is uniquely sensitive to treatment with all-trans retinoic acid (ATRA), which results in the expression of genes that induce the terminal granulocytic differentiation of the leukemic blasts. Here we report the identification of two ATRA responsive genes in APL cells, ID1 and ID2. These proteins act as antagonists of basic helix-loop-helix (bHLH) transcription factors. ATRA induced a rapid increase in ID1 and ID2, both in the APL cell line NB4 as well as in primary patient cells. In addition, a strong downregulation of E2A was observed. E2A acts as a general heterodimerization partner for many bHLH proteins that are involved in differentiation control in various tissues. The simultaneous upregulation of ID1 and ID2, and the downregulation of E2A suggest a role for bHLH proteins in the induction of differentiation of APL cells following ATRA treatment. To test the relevance of this upregulation, ID1 and ID2 were overexpressed in NB4 cells. Overexpression inhibited proliferation and induced a G0/G1 accumulation. These results indicate that ID1 and ID2 are important retinoic acid responsive genes in APL, and suggest that the inhibition of specific bHLH transcription factor complexes may play a role in the therapeutic effect of ATRA in APL.


Subject(s)
DNA-Binding Proteins , G1 Phase/drug effects , Leukemia, Promyelocytic, Acute/genetics , Repressor Proteins , Resting Phase, Cell Cycle/drug effects , Transcription Factors , Tretinoin/pharmacology , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Clone Cells/drug effects , Colony-Forming Units Assay , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Repressor Proteins/drug effects , Repressor Proteins/genetics , Repressor Proteins/pharmacology , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...