Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Blood ; 142(1): 90-105, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37146239

ABSTRACT

RNA-binding proteins (RBPs) form a large and diverse class of factors, many members of which are overexpressed in hematologic malignancies. RBPs participate in various processes of messenger RNA (mRNA) metabolism and prevent harmful DNA:RNA hybrids or R-loops. Here, we report that PIWIL4, a germ stem cell-associated RBP belonging to the RNase H-like superfamily, is overexpressed in patients with acute myeloid leukemia (AML) and is essential for leukemic stem cell function and AML growth, but dispensable for healthy human hematopoietic stem cells. In AML cells, PIWIL4 binds to a small number of known piwi-interacting RNA. Instead, it largely interacts with mRNA annotated to protein-coding genic regions and enhancers that are enriched for genes associated with cancer and human myeloid progenitor gene signatures. PIWIL4 depletion in AML cells downregulates the human myeloid progenitor signature and leukemia stem cell (LSC)-associated genes and upregulates DNA damage signaling. We demonstrate that PIWIL4 is an R-loop resolving enzyme that prevents R-loop accumulation on a subset of AML and LSC-associated genes and maintains their expression. It also prevents DNA damage, replication stress, and activation of the ATR pathway in AML cells. PIWIL4 depletion potentiates sensitivity to pharmacological inhibition of the ATR pathway and creates a pharmacologically actionable dependency in AML cells.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , Hematopoietic Stem Cells/metabolism , Cell Proliferation , Genomics , RNA, Messenger/metabolism , Neoplastic Stem Cells/pathology
2.
Blood Adv ; 7(3): 351-364, 2023 02 14.
Article in English | MEDLINE | ID: mdl-35468619

ABSTRACT

NPM1 is among the most frequently mutated genes in acute myeloid leukemia (AML). Mutations in the NPM1 gene result in the increased export of NPM1 to the cytoplasm (NPM1c) and are associated with multiple transforming events including the aberrant upregulation of MEIS1 that maintains stem cell and cell cycle-associated pathways in NPM1c AML. However, another consequence of the NPM1c mutation is the inadequate levels of NPM1 wild-type in the nucleus and nucleolus, caused by the loss of one wild-type allele in addition to enforced NPM1 nuclear export. The contribution of NPM1 haploinsufficiency independently of the NPM1 mutation to AML development and its relationship with MEIS1 function is poorly understood. Using mouse models, our study shows that NPM1 haploinsufficiency paired with MEIS1 overexpression is sufficient to induce a fully penetrant AML in mice that transcriptionally resembles human NPM1c AML. NPM1 haploinsufficiency alters MEIS1-binding occupancies such that it binds the promoter of the oncogene structural maintenance of chromosome protein 4 (SMC4) in NPM1 haploinsufficient AML cells but not in NPM1 wild-type-harboring Hoxa9/Meis1-transformed cells. SMC4 is higher expressed in haploinsufficient and NPM1c+ AML cells, which are more vulnerable to the disruption of the MEIS1-SMC4 axis compared with AML cells with nonmutated NPM1. Taken together, our study underlines that NPM1 haploinsufficiency on its own is a key factor of myeloid leukemogenesis and characterizes the MEIS1-SMC4 axis as a potential therapeutic target in this AML subtype.


Subject(s)
Haploinsufficiency , Leukemia, Myeloid, Acute , Humans , Animals , Mice , Leukemia, Myeloid, Acute/drug therapy , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Cell Nucleus/metabolism , Mutation , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/therapeutic use
3.
Leukemia ; 36(2): 416-425, 2022 02.
Article in English | MEDLINE | ID: mdl-34462525

ABSTRACT

Acute myeloid leukemia (AML) is considered a poor prognosis malignancy where patients exhibit altered glucose metabolism and stem cell signatures that contribute to AML growth and maintenance. Here, we report that the epigenetic factor, Ten-Eleven Translocation 3 (TET3) dioxygenase is overexpressed in AML patients and functionally validated human leukemic stem cells (LSCs), is required for leukemic growth by virtue of its regulation of glucose metabolism in AML cells. In human AML cells, TET3 maintains 5-hydroxymethylcytosine (5hmC) epigenetic marks and expression of early myeloid progenitor program, critical glucose metabolism and STAT5A signaling pathway genes, which also positively correlate with TET3 expression in AML patients. Consequently, TET3 depletion impedes hexokinase activity and L-Lactate production in AML cells. Conversely, overexpression of TET3 in healthy human hematopoietic stem progenitors (HSPCs) upregulates the expression of glucose metabolism, STAT5A signaling and AML associated genes, and impairs normal HSPC lineage differentiation in vitro. Finally, TET3 depletion renders AML cells highly sensitive to blockage of the TET3 downstream pathways glycolysis and STAT5 signaling via the combination of 2-Deoxy-D-glucose and STAT5 inhibitor which preferentially targets AML cells but spares healthy CD34+ HSPCs.


Subject(s)
Dioxygenases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Glucose/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Cell Proliferation , Dioxygenases/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 13(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669329

ABSTRACT

CXCR4 expression and downstream signaling have been identified as key factors in malignant hematopoiesis. Thus, up to 40% of all patients with Waldenström's macroglobulinemia (WM) carry an activating mutation of CXCR4 that leads to a more aggressive clinical course and inferior outcome upon treatment with the Bruton's tyrosine kinase inhibitor ibrutinib. Nevertheless, little is known about physiological mechanisms counteracting CXCR4 signaling in hematopoietic neoplasms. Recently, the endogenous human peptide EPI-X4 was identified as a natural CXCR4 antagonist that effectively blocks CXCL12-mediated receptor internalization and suppresses the migration and invasion of cancer cells towards a CXCL12 gradient. Here, we demonstrate that EPI-X4 efficiently binds to CXCR4 of WM cells and decreases their migration towards CXCL12. The CXCR4 inhibitory activity of EPI-X4 is accompanied by reduced expression of genes involved in MAPK signaling and energy metabolism. Notably, the anti-WM activity of EPI-X4 could be further augmented by the rational design of EPI-X4 derivatives showing higher binding affinity to CXCR4. In summary, these data demonstrate that a naturally occurring anti-CXCR4 peptide is able to interfere with WM cell behaviour, and that optimized derivatives of EPI-X4 may represent a promising approach in suppressing growth promoting CXCR4 signaling in WM.

5.
Leukemia ; 35(2): 389-403, 2021 02.
Article in English | MEDLINE | ID: mdl-32409690

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer characterized by skewed epigenetic patterns, raising the possibility of therapeutically targeting epigenetic factors in this disease. Here we report that among different cancer types, epigenetic factor TET1 is highly expressed in T-ALL and is crucial for human T-ALL cell growth in vivo. Knockout of TET1 in mice and knockdown in human T cell did not perturb normal T-cell proliferation, indicating that TET1 expression is dispensable for normal T-cell growth. The promotion of leukemic growth by TET1 was dependent on its catalytic property to maintain global 5-hydroxymethylcytosine (5hmC) marks, thereby regulate cell cycle, DNA repair genes, and T-ALL associated oncogenes. Furthermore, overexpression of the Tet1-catalytic domain was sufficient to augment global 5hmC levels and leukemic growth of T-ALL cells in vivo. We demonstrate that PARP enzymes, which are highly expressed in T-ALL patients, participate in establishing H3K4me3 marks at the TET1 promoter and that PARP1 interacts with the TET1 protein. Importantly, the growth related role of TET1 in T-ALL could be antagonized by the clinically approved PARP inhibitor Olaparib, which abrogated TET1 expression, induced loss of 5hmC marks, and antagonized leukemic growth of T-ALL cells, opening a therapeutic avenue for this disease.


Subject(s)
DNA Methylation , DNA-Binding Proteins/physiology , Gene Expression Regulation, Leukemic , Mixed Function Oxygenases/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Animals , Apoptosis , Cell Proliferation , Histones , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mixed Function Oxygenases/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Sci Rep ; 10(1): 371, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31941913

ABSTRACT

Acute myeloid leukemia (AML) is characterized by relapse and treatment resistance in a major fraction of patients, underlining the need of innovative AML targeting therapies. Here we analysed the therapeutic potential of an innovative biohybrid consisting of the tumor-associated peptide somatostatin and the photosensitizer ruthenium in AML cell lines and primary AML patient samples. Selective toxicity was analyzed by using CD34 enriched cord blood cells as control. Treatment of OCI AML3, HL60 and THP1 resulted in a 92, and 99 and 97% decrease in clonogenic growth compared to the controls. Primary AML cells demonstrated a major response with a 74 to 99% reduction in clonogenicity in 5 of 6 patient samples. In contrast, treatment of CD34+ CB cells resulted in substantially less reduction in colony numbers. Subcellular localization assays of RU-SST in OCI-AML3 cells confirmed strong co-localization of RU-SST in the lysosomes compared to the other cellular organelles. Our data demonstrate that conjugation of a Ruthenium complex with somatostatin is efficiently eradicating LSC candidates of patients with AML. This indicates that receptor mediated lysosomal accumulation of photodynamic metal complexes is a highly attractive approach for targeting AML cells.


Subject(s)
Leukemia, Myeloid, Acute/therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Receptors, Somatostatin/metabolism , Ruthenium/therapeutic use , Somatostatin/therapeutic use , Adult , Aged , Apoptosis , Cell Line, Tumor , Drug Stability , Female , Fetal Blood/metabolism , Humans , Lysosomes/metabolism , Male , Middle Aged , Photosensitizing Agents/chemistry , Reactive Oxygen Species/metabolism
8.
Leukemia ; 34(4): 1027-1037, 2020 04.
Article in English | MEDLINE | ID: mdl-31758089

ABSTRACT

Acute myeloid leukemia (AML) is the most common acute leukemia in adults and is propagated by leukemic stem cells (LSCs), often characterized by deregulated Wnt signaling. We previously showed that the central transcriptional mediator of Wnt signaling LEF1 is able to cause AML in mice and acts as an independent prognostic factor in normal karyotype AML. Here, we show that treatment naïve normal karyotype AML as well as samples AML LSCs predominantly express the long ß-catenin-binding isoform of LEF1 in sharp contrast to normal human hematopoietic stem cells, which lack expression of the long isoform, but express the short N-terminally truncated isoform with loss of the ß-catenin-binding site. Gene expression and ChiP-Seq analyses in mice linked the long isoform to Wnt-ß-catenin signaling and oncogenic pathways, the N-terminally truncated isoform to stemness associated genes. Approaches impairing binding of LEF1 to ß-catenin significantly impaired AML growth, but spared normal hematopoietic stem cells. This report now demonstrates a striking difference of LEF1 isoform expression between normal and AML cells, contributing to higher vulnerability of leukemic cells to approaches targeting ß-catenin/LEF1 interaction.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mutation , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor , Carcinogenesis , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplastic Stem Cells/metabolism , Protein Isoforms , Wnt Signaling Pathway , beta Catenin
9.
Blood Adv ; 3(22): 3729-3739, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31770439

ABSTRACT

Acute erythroid leukemia (AEL) is a rare and aggressive form of acute leukemia, the biology of which remains poorly understood. Here we demonstrate that the ParaHox gene CDX4 is expressed in patients with acute erythroid leukemia, and that aberrant expression of Cdx4 induced homogenously a transplantable acute erythroid leukemia in mice. Gene expression analyses demonstrated upregulation of genes involved in stemness and leukemogenesis, with parallel downregulation of target genes of Gata1 and Gata2 responsible for erythroid differentiation. Cdx4 induced a proteomic profile that overlapped with a cluster of proteins previously defined to represent the most primitive human erythroid progenitors. Whole-exome sequencing of diseased mice identified recurrent mutations significantly enriched for transcription factors involved in erythroid lineage specification, as well as TP53 target genes partly identical to the ones reported in patients with AEL. In summary, our data indicate that Cdx4 is able to induce stemness and inhibit terminal erythroid differentiation, leading to the development of AEL in association with co-occurring mutations.


Subject(s)
Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Adult , Aged , Animals , Biomarkers, Tumor , Cell Differentiation/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Association Studies , Hematopoiesis/genetics , Humans , Immunophenotyping , Male , Mice , Middle Aged , Mutation , Whole Genome Sequencing
10.
Oncotarget ; 7(52): 86889-86901, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27888632

ABSTRACT

Homeobox genes are key regulators in normal and malignant hematopoiesis. The human Vent-like homeobox gene VENTX, a putative homolog of the Xenopus laevis Xvent-2 gene, was shown to be highly expressed in normal myeloid cells and in patients with acute myeloid leukemia. We now demonstrate that constitutive expression of VENTX suppresses expression of genes responsible for terminal erythroid differentiation in normal CD34+ stem and progenitor cells. Transplantation of bone marrow progenitor cells retrovirally engineered to express VENTX caused massive expansion of primitive erythroid cells and partly acute erythroleukemia in transplanted mice. The leukemogenic potential of VENTX was confirmed in the AML1-ETO transplantation model, as in contrast to AML1-ETO alone co-expression of AML1-ETO and VENTX induced acute myeloid leukemia, partly expressing erythroid markers, in all transplanted mice. VENTX was highly expressed in patients with primary human erythroleukemias and knockdown of VENTX in the erythroleukemic HEL cell line significantly blocked cell growth. In summary, these data indicate that VENTX is able to perturb erythroid differentiation and to contribute to myeloid leukemogenesis when co-expressed with appropriate AML oncogenes and point to its potential significance as a novel therapeutic target in AML.


Subject(s)
Cell Proliferation/genetics , Erythroid Cells/metabolism , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation, Leukemic , Homeodomain Proteins/metabolism , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Myeloid, Acute/metabolism , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Middle Aged , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , RNA Interference
11.
Cell Rep ; 16(2): 498-507, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346355

ABSTRACT

Homeobox genes are known to be key factors in leukemogenesis. Although the TALE family homeodomain factor Meis1 has been linked to malignancy, a role for MEIS2 is less clear. Here, we demonstrate that MEIS2 is expressed at high levels in patients with AML1-ETO-positive acute myeloid leukemia and that growth of AML1-ETO-positive leukemia depends on MEIS2 expression. In mice, MEIS2 collaborates with AML1-ETO to induce acute myeloid leukemia. MEIS2 binds strongly to the Runt domain of AML1-ETO, indicating a direct interaction between these transcription factors. High expression of MEIS2 impairs repressive DNA binding of AML1-ETO, inducing increased expression of genes such as the druggable proto-oncogene YES1. Collectively, these data describe a pivotal role for MEIS2 in AML1-ETO-induced leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Transcription Factors/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression , Gene Expression Regulation, Leukemic , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplasm Transplantation , Oncogene Proteins, Fusion/metabolism , Oncogenes , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription Factors/metabolism
12.
Stem Cells ; 30(12): 2603-11, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22996918

ABSTRACT

Piwi proteins and their associated piRNAs are essential for preserving the self-renewal property of mammalian germ stem cells. Their highly conserved role in CpG island DNA methylation and chromatin modifications in germ stem cells has long been associated with transposon silencing but recent reports hint at protein coding regions being targets for Piwi-mediated epigenetic changes as well. Interestingly, the expression of PIWI family members is not restricted to the germline, and certain members have also been implicated in tumorigenesis in cases of adenocarcinomas, gliomas, and sarcomas. The following review discusses our knowledge of the function of Piwi proteins and piRNAs in suppressing transposable elements while maintaining the self-renewing population of germ stem cells. We also highlight the somatic function of Piwi as an epigenetic modifier. Furthermore, we summarize the recently uncovered involvement of Piwi proteins and piRNAs in various cancers.


Subject(s)
Argonaute Proteins/physiology , DNA Transposable Elements , RNA, Small Interfering/physiology , Stem Cells/physiology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Gene Silencing , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stem Cells/cytology , Stem Cells/metabolism
13.
Blood ; 120(10): 2118-26, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22815426

ABSTRACT

Lymphoid enhancer-binding factor-1 (LEF1) is a key transcription factor of Wnt signaling. We recently showed that aberrant LEF1 expression induces acute myeloid leukemia (AML) in mice, and found high LEF1 expression in a subset of cytogenetically normal AML (CN-AML) patients. Whether LEF1 expression associates with clinical and molecular patient characteristics and treatment outcomes remained unknown. We therefore studied LEF1 expression in 210 adults with CN-AML treated on German AML Cooperative Group trials using microarrays. High LEF1 expression (LEF1high) associated with significantly better relapse-free survival (RFS; P < .001), overall survival (OS; P < .001), and event-free survival (EFS; P < .001). In multivariable analyses adjusting for established prognosticators, LEF1high status remained associated with prolonged RFS (P = .007), OS (P = .01), and EFS (P = .003). In an independent validation cohort of 196 CN-AML patients provided by the German-Austrian AML Study Group, LEF1high patients had significantly longer OS (P = .02) and EFS (P = .04). We validated the prognostic relevance of LEF1 expression by quantitative PCR, thereby providing a clinically applicable platform to incorporate this marker into future risk-stratification systems for CN-AML. Gene-expression profiling and immunophenotyping revealed up-regulation of lymphopoiesis-related genes and lymphoid cell-surface antigens in LEF1high patients. In summary, we provide evidence that high LEF1 expression is a novel favorable prognostic marker in CN-AML.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Gene Expression , Leukemia, Myeloid, Acute/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Female , Gene Expression Profiling , Humans , Immunophenotyping , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Survival Analysis , Wnt Signaling Pathway/genetics
14.
Blood ; 120(3): 519-27, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22547580

ABSTRACT

During the past decade it was recognized that homeobox gene families such as the clustered Hox genes play pivotal roles both in normal and malignant hematopoiesis. More recently, similar roles have also become apparent for members of the ParaHox gene cluster, evolutionarily closely related to the Hox gene cluster. This is in particular found for the caudal-type homeobox genes (Cdx) genes, known to act as upstream regulators of Hox genes. The CDX gene family member CDX2 belongs to the most frequent aberrantly expressed proto-oncogenes in human acute leukemias and is highly leukemogenic in experimental models. Correlative studies indicate that CDX2 functions as master regulator of perturbed HOX gene expression in human acute myeloid leukemia, locating this ParaHox gene at a central position for initiating and maintaining HOX gene dysregulation as a driving leukemogenic force. There are still few data about potential upstream regulators initiating aberrant CDX2 expression in human leukemias or about critical downstream targets of CDX2 in leukemic cells. Characterizing this network will hopefully open the way to therapeutic approaches that target deregulated ParaHox genes in human leukemia.


Subject(s)
Gene Expression Regulation, Leukemic , Genes, Homeobox/genetics , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Humans
15.
Exp Hematol ; 39(3): 272-81, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21168469

ABSTRACT

Stem cells are defined as cells that have the ability to perpetuate themselves through self-renewal and to generate functional mature cells by differentiation. During each stage, coordinated gene expression is crucial to maintain the balance between self-renewal and differentiation. Disturbance of this accurately balanced system can lead to a variety of malignant disorders. In mammals, DNA cytosine-5 methylation is a well-studied epigenetic pathway that is catalyzed by DNA methyltransferases and is implicated in the control of balanced gene expression, but also in hematological malignancies. In this review, we focus on the TET (ten-eleven-translocation) genes, which recently were identified to catalyze the conversion of cytosine-5 methylation to 5-hydroxymethyl-cytosine, an intermediate form potentially involved in demethylation. In addition, members of the TET family are playing a role in ES cell maintenance and inner cell mass cell specification and were demonstrated to be involved in hematological malignancies. Recently, a correlation between low genomic 5-hydroxymethyl-cytosine and TET2 mutation status was shown in patients with myeloid malignancies.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Hematologic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Dioxygenases , Embryonic Stem Cells/pathology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Mutation , Proto-Oncogene Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 107(39): 16946-51, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20833819

ABSTRACT

Recent data indicate that a variety of regulatory molecules active in embryonic development may also play a role in the regulation of early hematopoiesis. Here we report that the human Vent-like homeobox gene VENTX, a putative homolog of the Xenopus xvent2 gene, is a unique regulatory hematopoietic gene that is aberrantly expressed in CD34(+) leukemic stem-cell candidates in human acute myeloid leukemia (AML). Quantitative RT-PCR documented expression of the gene in lineage positive hematopoietic subpopulations, with the highest expression in CD33(+) myeloid cells. Notably, expression levels of VENTX were negligible in normal CD34(+)/CD38(-) or CD34(+) human progenitor cells. In contrast to this, leukemic CD34(+)/CD38(-) cells from AML patients with translocation t(8,21) and normal karyotype displayed aberrantly high expression of VENTX. Gene expression and pathway analysis demonstrated that in normal CD34(+) cells enforced expression of VENTX initiates genes associated with myeloid development and down-regulates genes involved in early lymphoid development. Functional analyses confirmed that aberrant expression of VENTX in normal CD34(+) human progenitor cells perturbs normal hematopoietic development, promoting generation of myeloid cells and impairing generation of lymphoid cells in vitro and in vivo. Stable knockdown of VENTX expression inhibited the proliferation of human AML cell lines. Taken together, these data extend our insights into the function of embryonic mesodermal factors in human postnatal hematopoiesis and indicate a role for VENTX in normal and malignant myelopoiesis.


Subject(s)
Gene Expression Regulation, Leukemic , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Myeloid Cells/cytology , Myelopoiesis/genetics , Coculture Techniques , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Myeloid Cells/metabolism
17.
J Exp Med ; 205(3): 515-22, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18316418

ABSTRACT

Canonical Wnt signaling is critically involved in normal hematopoietic development and the self-renewal process of hematopoietic stem cells (HSCs). Deregulation of this pathway has been linked to a large variety of cancers, including different subtypes of leukemia. Lef-1 is a major transcription factor of this pathway and plays a pivotal role in lymphoid differentiation as well as in granulopoiesis. Here, we demonstrate Lef-1 expression in murine HSCs as well as its expression in human leukemia. Mice transplanted with bone marrow retrovirally transduced to express Lef-1 or a constitutive active Lef-1 mutant showed a severe disturbance of normal hematopoietic differentiation and finally developed B lymphoblastic and acute myeloid leukemia (AML). Lef-1-induced AMLs were characterized by immunoglobulin (Ig) DH-JH rearrangements and a promiscuous expression of lymphoid and myeloid regulatory factors. Furthermore, single cell experiments and limiting dilution transplantation assays demonstrated that Lef-1-induced AML was propagated by a leukemic stem cell with lymphoid characteristics displaying Ig DH-JH rearrangements and a B220(+) myeloid marker(-) immunophenotype. These data indicate a thus far unknown role of Lef-1 in the biology of acute leukemia, pointing to the necessity of balanced Lef-1 expression for an ordered hematopoietic development.


Subject(s)
Leukemia/etiology , Lymphoid Enhancer-Binding Factor 1/metabolism , Wnt Proteins/metabolism , Animals , Gene Expression , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/genetics , Leukemia/metabolism , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
18.
Blood ; 111(1): 309-19, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17855634

ABSTRACT

The mechanisms underlying deregulation of HOX gene expression in AML are poorly understood. The ParaHox gene CDX2 was shown to act as positive upstream regulator of several HOX genes. In this study, constitutive expression of Cdx2 caused perturbation of leukemogenic Hox genes such as Hoxa10 and Hoxb8 in murine hematopoietic progenitors. Deletion of the N-terminal domain of Cdx2 abrogated its ability to perturb Hox gene expression and to cause acute myeloid leukemia (AML) in mice. In contrast inactivation of the putative Pbx interacting site of Cdx2 did not change the leukemogenic potential of the gene. In an analysis of 115 patients with AML, expression levels of CDX2 were closely correlated with deregulated HOX gene expression. Patients with normal karyotype showed a 14-fold higher expression of CDX2 and deregulated HOX gene expression compared with patients with chromosomal translocations such as t(8:21) or t(15;17). All patients with AML with normal karyotype tested were negative for CDX1 and CDX4 expression. These data link the leukemogenic potential of Cdx2 to its ability to dysregulate Hox genes. They furthermore correlate the level of CDX2 expression with HOX gene expression in human AML and support a potential role of CDX2 in the development of human AML with aberrant Hox gene expression.


Subject(s)
Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Transcription Factors/genetics , Adult , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , CDX2 Transcription Factor , Cell Line, Tumor , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Karyotyping , Mice , Mutagenesis , NIH 3T3 Cells , Protein Structure, Tertiary , Transcription Factors/chemistry , Transcription Factors/metabolism , Translocation, Genetic , Up-Regulation/physiology
19.
Cancer Cell ; 10(5): 363-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17097559

ABSTRACT

A challenge for the development of therapies selectively targeting leukemic stem cells in acute myeloid leukemia (AML) is their similarity to normal hematopoietic stem cells (HSCs). Here we demonstrate that the leukemia-propagating cell in murine CALM/AF10-positive AML differs from normal HSCs by B220 surface expression and immunoglobulin heavy chain rearrangement. Furthermore, depletion of B220+ cells in leukemic transplants impaired development of leukemia in recipients. As in the murine model, human CALM/AF10-positive AML was characterized by CD45RA (B220)-positive, IG DH-JH rearranged leukemic cells. These data demonstrate in a murine leukemia model that AML can be propagated by a transformed progenitor with lymphoid characteristics, which can be targeted by antibodies that do not crossreact with normal HSCs.


Subject(s)
Disease Models, Animal , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/physiopathology , Monomeric Clathrin Assembly Proteins/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Transplantation , Cell Transformation, Neoplastic , Humans , Leukocyte Common Antigens/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Mice , Monomeric Clathrin Assembly Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Survival Rate
20.
J Clin Invest ; 115(8): 2159-68, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025155

ABSTRACT

The molecular characterization of leukemia has demonstrated that genetic alterations in the leukemic clone frequently fall into 2 classes, those affecting transcription factors (e.g., AML1-ETO) and mutations affecting genes involved in signal transduction (e.g., activating mutations of FLT3 and KIT). This finding has favored a model of leukemogenesis in which the collaboration of these 2 classes of genetic alterations is necessary for the malignant transformation of hematopoietic progenitor cells. The model is supported by experimental data indicating that AML1-ETO and FLT3 length mutation (FLT3-LM), 2 of the most frequent genetic alterations in AML, are both insufficient on their own to cause leukemia in animal models. Here we report that AML1-ETO collaborates with FLT3-LM in inducing acute leukemia in a murine BM transplantation model. Moreover, in a series of 135 patients with AML1-ETO-positive AML, the most frequently identified class of additional mutations affected genes involved in signal transduction pathways including FLT3-LM or mutations of KIT and NRAS. These data support the concept of oncogenic cooperation between AML1-ETO and a class of activating mutations, recurrently found in patients with t(8;21), and provide a rationale for therapies targeting signal transduction pathways in AML1-ETO-positive leukemias.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Translocation, Genetic , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Core Binding Factor Alpha 2 Subunit , Disease Models, Animal , Female , Genes, ras/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukopoiesis/genetics , Male , Mice , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RUNX1 Translocation Partner 1 Protein , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...