Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(15)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34359759

ABSTRACT

We recently characterised the NUP98-HOXD13 (NHD13) mouse as a model of T-cell pre-leukaemia, featuring thymocytes that can engraft in recipient animals and progress to T-cell acute lymphoblastic leukaemia (T-ALL). However, loss of this engraftment ability by deletion of Lyl1 did not result in any loss of leukemogenesis activity. In the present study, we observe that NHD13 thymocytes overexpress EPHA3, and we characterise thymocyte behaviour in NHD13 mice with deletion of EphA3, which show a markedly reduced incidence of T-ALL. Deletion of EphA3 from the NHD13 mice does not prevent the abnormal accumulation or transplantation ability of these thymocytes. However, upon transplantation, these cells are unable to block the normal progression of recipient wild type (WT) progenitor cells through the normal developmental pathway. This is in contrast to the EphA3+/+ NHD13 thymocytes, which block the progression of incoming WT progenitors past the DN1 stage. Therefore, EphA3 is not critical for classical self-renewal, but is essential for mediating an interaction between the abnormally self-renewing cells and healthy progenitors-an interaction that results in a failure of the healthy cells to differentiate normally. We speculate that this may orchestrate a loss of healthy cell competition, which in itself has been demonstrated to be oncogenic, and that this may explain the decrease in T-ALL incidence in the absence of EphA3. We suggest that pre-leukaemic self-renewal in this model is a complex interplay of cell-intrinsic and -extrinsic factors, and that multiple redundant pathways to leukaemogenesis are active.

2.
Cancers (Basel) ; 13(6)2021 Mar 18.
Article in English | MEDLINE | ID: mdl-33803727

ABSTRACT

Pre-leukemia is a catch-all term for any haematological condition which predisposes the individual towards developing leukemia [...].

4.
Leukemia ; 33(8): 1868-1880, 2019 08.
Article in English | MEDLINE | ID: mdl-30700838

ABSTRACT

T cell acute lymphoblastic leukaemia (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. While it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukaemia (AML) as well as T-ALL. We find that thymocytes from preleukaemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional programme closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression programme in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus multiple T cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1; however, NHD13 can also promote T-ALL via an alternative pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Homeodomain Proteins/genetics , LIM Domain Proteins/physiology , Neoplasm Proteins/physiology , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymocytes/physiology , Transcription Factors/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Growth Factors ; 32(6): 176-89, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25391995

ABSTRACT

Eph receptor tyrosine kinases control cell-cell interactions during normal and oncogenic development, and are implicated in a range of processes including angiogenesis, stem cell maintenance and metastasis. They are thus of great interest as targets for cancer therapy. EphA3, originally isolated from leukemic and melanoma cells, is presently one of the most promising therapeutic targets, with multiple tumor-promoting roles in a variety of cancer types. This review focuses on EphA3, its functions in controlling cellular behavior, both in normal and pathological development, and most particularly in cancer.


Subject(s)
Neoplasms/metabolism , Receptors, Eph Family/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/chemistry , Receptors, Eph Family/genetics
6.
Leuk Res ; 37(12): 1668-73, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24090997

ABSTRACT

NUP98 is among the most promiscuously translocated genes in hematological diseases. Among the 28 known fusion partners, there are two categories: homeobox genes and non-homeobox genes. The homeobox fusion partners are well-studied in animal models, resulting in HoxA cluster overexpression and hematological disease. The non-homeobox fusion partners are less well studied. We created transgenic animal models for three NUP98 fusion genes (one homeobox, two non-homeobox), and show that in this system, the NUP98-homeobox fusion promotes self-renewal and aberrant gene expression to a significantly greater extent. We conclude that homeobox partners create more potent NUP98 fusion oncogenes than do non-homeobox partners.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/physiology , Translocation, Genetic/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , DNA Topoisomerases, Type I/genetics , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Exp Hematol ; 40(12): 1016-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22885519

ABSTRACT

NUP98-HOXD13 (NHD13) and CALM-AF10 (CA10) are oncogenic fusion proteins produced by recurrent chromosomal translocations in patients with acute myeloid leukemia (AML). Transgenic mice that express these fusions develop AML with a long latency and incomplete penetrance, suggesting that collaborating genetic events are required for leukemic transformation. We employed genetic techniques to identify both preleukemic abnormalities in healthy transgenic mice as well as collaborating events leading to leukemic transformation. Candidate gene resequencing revealed that 6 of 27 (22%) CA10 AMLs spontaneously acquired a Ras pathway mutation and 8 of 27 (30%) acquired an Flt3 mutation. Two CA10 AMLs acquired an Flt3 internal-tandem duplication, demonstrating that these mutations can be acquired in murine as well as human AML. Gene expression profiles revealed a marked upregulation of Hox genes, particularly Hoxa5, Hoxa9, and Hoxa10 in both NHD13 and CA10 mice. Furthermore, mir196b, which is embedded within the Hoxa locus, was overexpressed in both CA10 and NHD13 samples. In contrast, the Hox cofactors Meis1 and Pbx3 were differentially expressed; Meis1 was increased in CA10 AMLs but not NHD13 AMLs, whereas Pbx3 was consistently increased in NHD13 but not CA10 AMLs. Silencing of Pbx3 in NHD13 cells led to decreased proliferation, increased apoptosis, and decreased colony formation in vitro, suggesting a previously unexpected role for Pbx3 in leukemic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Leukemia/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Animals , Apoptosis/genetics , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Genetic Loci , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics
8.
Blood ; 120(12): 2475-83, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22855610

ABSTRACT

Programmed cell death or apoptosis is a prominent feature of low-risk myelodysplastic syndromes (MDS), although the underlying mechanism remains controversial. High-risk MDS have less apoptosis associated with increased expression of the prosurvival BCL2-related proteins. To address the mechanism and pathogenic role of apoptosis and BCL2 expression in MDS, we used a mouse model resembling human MDS, in which the fusion protein NUP98-HOXD13 (NHD13) of the chromosomal translocation t(2;11)(q31;p15) is expressed in hematopoietic cells. Hematopoietic stem and progenitor cells from 3-month-old mice had increased rates of apoptosis associated with increased cell cycling and DNA damage. Gene expression profiling of these MDS progenitors revealed a specific reduction in Bcl2. Restoration of Bcl2 expression by a BCL2 transgene blocked apoptosis of the MDS progenitors, which corrected the macrocytic anemia. Blocking apoptosis also restored cell-cycle quiescence and reduced DNA damage in the MDS progenitors. We expected that preventing apoptosis would accelerate malignant transformation to acute myeloid leukemia (AML). However, contrary to expectations, preventing apoptosis of premalignant cells abrogated transformation to AML. In contrast to the current dogma that overcoming apoptosis is an important step toward cancer, this work demonstrates that gaining a survival advantage of premalignant cells may delay or prevent leukemic progression.


Subject(s)
Apoptosis , Cell Transformation, Neoplastic/pathology , Hematopoietic Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Blood ; 119(12): 2883-94, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22323452

ABSTRACT

Constitutive activation of FLT3 by internal tandem duplication (ITD) is one of the most common molecular alterations in acute myeloid leukemia (AML). FLT3/ITD mutations have also been observed in myelodysplastic syndrome patients both before and during progression to AML. Previous work has shown that insertion of an FLT3/ITD mutation into the murine Flt3 gene induces a myeloproliferative neoplasm, but not progression to acute leukemia, suggesting that additional cooperating events are required. We therefore combined the FLT3/ITD mutation with a model of myelodysplastic syndrome involving transgenic expression of the Nup98-HoxD13 (NHD13) fusion gene. Mice expressing both the FLT3/ITD and NHD13 transgene developed AML with 100% penetrance and short latency. These leukemias were driven by mutant FLT3 expression and were susceptible to treatment with FLT3 tyrosine kinase inhibitors. We also observed a spontaneous loss of the wild-type Flt3 allele in these AMLs, further modeling the loss of the heterozygosity phenomenon that is seen in human AML with FLT3-activating mutations. Because resistance to FLT3 inhibitors remains an important clinical issue, this model may help identify new molecular targets in collaborative signaling pathways.


Subject(s)
Disease Models, Animal , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Blotting, Western , Flow Cytometry , Gene Knock-In Techniques , Humans , Mice , Mice, Transgenic , Mutation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Blood ; 118(24): 6247-57, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21948299

ABSTRACT

Structural chromosomal rearrangements of the Nucleoporin 98 gene (NUP98), primarily balanced translocations and inversions, are associated with a wide array of hematopoietic malignancies. NUP98 is known to be fused to at least 28 different partner genes in patients with hematopoietic malignancies, including acute myeloid leukemia, chronic myeloid leukemia in blast crisis, myelodysplastic syndrome, acute lymphoblastic leukemia, and bilineage/biphenotypic leukemia. NUP98 gene fusions typically encode a fusion protein that retains the amino terminus of NUP98; in this context, it is important to note that several recent studies have demonstrated that the amino-terminal portion of NUP98 exhibits transcription activation potential. Approximately half of the NUP98 fusion partners encode homeodomain proteins, and at least 5 NUP98 fusions involve known histone-modifying genes. Several of the NUP98 fusions, including NUP98-homeobox (HOX)A9, NUP98-HOXD13, and NUP98-JARID1A, have been used to generate animal models of both lymphoid and myeloid malignancy; these models typically up-regulate HOXA cluster genes, including HOXA5, HOXA7, HOXA9, and HOXA10. In addition, several of the NUP98 fusion proteins have been shown to inhibit differentiation of hematopoietic precursors and to increase self-renewal of hematopoietic stem or progenitor cells, providing a potential mechanism for malignant transformation.


Subject(s)
Hematologic Neoplasms/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Hematologic Neoplasms/metabolism , Homeodomain Proteins/genetics , Humans , Nuclear Pore/physiology , Nuclear Pore Complex Proteins/physiology , Sequence Inversion , Transcription, Genetic , Translocation, Genetic
11.
Blood ; 115(6): 1194-203, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20007546

ABSTRACT

The t(10;11) translocation results in a CALM-AF10 fusion gene in a subset of leukemia patients. Expression of a CALM-AF10 transgene results in leukemia, with prolonged latency and incomplete penetrance, suggesting that additional events are necessary for leukemic transformation. CALM-AF10 mice infected with the MOL4070LTR retrovirus developed acute leukemia, and ligation-mediated polymerase chain reaction was used to identify retroviral insertions at 19 common insertion sites, including Zeb2, Nf1, Mn1, Evi1, Ift57, Mpl, Plag1, Kras, Erg, Vav1, and Gata1. A total of 26% (11 of 42) of the mice had retroviral integrations near Zeb2, a transcriptional corepressor leading to overexpression of the Zeb2-transcript. A total of 91% (10 of 11) of mice with Zeb2 insertions developed B-lineage acute lymphoblastic leukemia, suggesting that Zeb2 activation promotes the transformation of CALM-AF10 hematopoietic precursors toward B-lineage leukemias. More than half of the mice with Zeb2 integrations also had Nf1 integrations, suggesting cooperativity among CALM-AF10, Zeb2, and Ras pathway mutations. We searched for Nras, Kras, and Ptpn11 point mutations in the CALM-AF10 leukemic mice. Three mutations were identified, all of which occurred in mice with Zeb2 integrations, consistent with the hypothesis that Zeb2 and Ras pathway activation promotes B-lineage leukemic transformation in concert with CALM-AF10.


Subject(s)
Homeodomain Proteins/metabolism , Leukemia, Experimental/genetics , Mutagenesis, Insertional , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Repressor Proteins/metabolism , Retroviridae/genetics , Animals , Blotting, Southern , Blotting, Western , Cell Proliferation , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Immunophenotyping , Leukemia, B-Cell/genetics , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Transgenic , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Zinc Finger E-box Binding Homeobox 2 , ras Proteins/genetics
12.
J Immunol ; 183(10): 6227-35, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841179

ABSTRACT

Expression of a NUP98-HOXD13 (NHD13) fusion gene leads to myelodysplastic syndrome in mice. In addition to ineffective hematopoiesis, we observed that NHD13 mice were lymphopenic; the lymphopenia was due to a decrease in both T and B lymphocytes. Although the pro-B cell (B220(+)/CD43(+)) populations from the NHD13 and wild-type mice were similar, the NHD13 mice showed decreased pre-B cells (B220(+)/CD43(-)), indicating impaired differentiation at the pro-B to pre-B stage. Thymi from NHD13 mice were smaller and overexpressed Hoxa cluster genes, including Hoxa7, Hoxa9, and Hoxa10. In addition, the NHD13 thymi contained fewer thymocytes, with an increased percentage of CD4(-)/CD8(-) (double-negative (DN)) cells and a decreased percentage of CD4(+)/CD8(+) (double-positive) cells; the DN1/DN2 population was increased and the DN3/DN4 population was decreased, suggesting a partial block at the DN2 to DN3 transition. To determine clonality of the thymocytes, we used degenerate RT-PCR to identify clonal Tcrb gene rearrangements. Five of six NHD13 thymi showed an unusual Tcrb gene rearrangement pattern with common, clonal DJ rearrangements, but distinct V-D junctions, suggesting a marked clonal expansion of thymocytes that had undergone a DJ rearrangement, but not completed a VDJ rearrangement. Taken together, these findings demonstrate that expression of the NHD13 transgene inhibits lymphoid as well as myeloid and erythroid differentiation, results in overexpression of Hoxa cluster genes, and leads to a precursor T cell lymphoblastic leukemia/lymphoma.


Subject(s)
B-Lymphocytes/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Homeodomain Proteins/metabolism , Myelodysplastic Syndromes/immunology , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression/genetics , Gene Expression/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/immunology , Oncogene Proteins, Fusion/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Transcription Factors/genetics , Transcription Factors/immunology
13.
Proc Natl Acad Sci U S A ; 105(37): 14088-93, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18768819

ABSTRACT

The myelodysplastic syndromes (MDS) comprise a group of premalignant hematologic disorders characterized by ineffective hematopoiesis, dysplasia, and transformation to acute myeloid leukemia (AML). Although it is well established that many malignancies can be transplanted, there is little evidence to demonstrate that a premalignant disease entity, such as MDS or colonic polyps, can be transplanted and subsequently undergo malignant transformation in vivo. Using mice that express a NUP98-HOXD13 (NHD13) transgene in hematopoietic tissues, we show that a MDS can be transplanted to WT recipients. Recipients of the MDS bone marrow displayed all of the critical features of MDS, including peripheral blood cytopenias, dysplasia, and transformation to AML. Even when transplanted with a 10-fold excess of WT cells, the NHD13 cells outcompeted the WT cells over a 38-week period. Limiting-dilution experiments demonstrated that the frequency of the cell that could transmit the disease was approximately 1/6,000-1/16,000 and that the MDS was also transferable to secondary recipients as a premalignant condition. Transformation to AML in primary transplant recipients was generally delayed (46-49 weeks after transplant); however, 6 of 10 secondary transplant recipients developed AML. These findings demonstrate that MDS originates in a transplantable, premalignant, long-term repopulating, MDS-initiating cell.


Subject(s)
Cell Lineage , Hematopoietic Stem Cell Transplantation , Hematopoietic System/cytology , Myelodysplastic Syndromes/pathology , Animals , Bone Marrow/immunology , Cell Separation , Leukemia/pathology , Mice , Mice, Inbred C57BL , Myelodysplastic Syndromes/surgery , Time Factors
14.
Haematologica ; 93(9): 1394-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18603548

ABSTRACT

Expression of a NUP98-HOXD13 (NHD13) fusion gene, initially identified in a patient with myelodysplastic syndrome, leads to a highly penetrant myelodysplastic syndrome in mice that recapitulates all of the key features of the human disease. Expansion of undifferentiated lineage negative (lin(neg)) hematopoietic precursors that express NHD13 was markedly inhibited (30-fold) in vitro. Decreased expansion was accompanied by decreased production of terminally differentiated cells, indicating impaired differentiation of NHD13 precursors. Rather than differentiate, the majority (80%) of NHD13 lin(neg) precursors underwent apoptotic cell death when induced to differentiate. These findings demonstrate that NHD13 lin(neg) cells provide a tractable in vitro system for studies of myelodysplastic syndrome.


Subject(s)
Apoptosis , Cell Differentiation , Hematopoietic System/cytology , Myelodysplastic Syndromes/pathology , Animals , Cell Line , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
J Natl Cancer Inst Monogr ; (39): 64-8, 2008.
Article in English | MEDLINE | ID: mdl-18648006

ABSTRACT

The myelodysplastic syndromes (MDS) are a group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias, dysplasia, and a propensity for transformation to acute myeloid leukemia (AML). A wide spectrum of genetic aberrations has been associated with MDS, including chromosomal translocations involving the NUP98 gene, most commonly leading to fusions of NUP98 with abd-b group HOX genes, including HOXD13. We used vav regulatory elements to direct expression of a NUP98-HOXD13 (NHD13) fusion gene in hematopoietic tissues. NHD13 transgenic mice faithfully recapitulate all the key features of MDS, including peripheral blood cytopenias, bone marrow dysplasia and apoptosis, and transformation to acute leukemia. The MDS that develops in NHD13 transgenic mice is highly lethal; within 14 months, 90% of the mice died of either leukemic transformation or severe anemia and leukopenia due to progressive MDS. These mice provide a preclinical model that can be used for the evaluation of MDS therapy and biology.


Subject(s)
Homeodomain Proteins/genetics , Myelodysplastic Syndromes/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Translocation, Genetic , Animals , Chromosome Aberrations , Disease Models, Animal , Humans , Mice , Mice, Transgenic
16.
Blood ; 112(5): 2017-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18566322

ABSTRACT

The NUP98-HOXD13 (NHD13) fusion gene occurs in patients with myelodysplastic syndrome (MDS) and acute nonlymphocytic leukemia (ANLL). We reported that transgenic mice expressing NHD13 develop MDS, and that more than half of these mice eventually progress to acute leukemia. The latency period suggests a requirement for at least 1 complementary event before leukemic transformation. We conducted a candidate gene search for complementary events focused on genes that are frequently mutated in human myeloid leukemia. We investigated 22 ANLL samples and found a high frequency of Nras and Kras mutations, an absence of Npm1, p53, Runx1, Kit and Flt3 mutations, and a single Cbl mutation. Our findings support a working hypothesis that predicts that ANLL cases have one mutation which inhibits differentiation, and a complementary mutation which enhances proliferation or inhibit apoptosis. In addition, we provide the first evidence for spontaneous collaborating mutations in a genetically engineered mouse model of ANLL.


Subject(s)
Genes, ras , Leukemia, Experimental/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-cbl/genetics , Animals , Base Sequence , Cell Transformation, Neoplastic/genetics , Cocarcinogenesis , DNA, Neoplasm/genetics , Disease Models, Animal , Humans , Leukemia, Experimental/etiology , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Transgenic , Models, Biological , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/genetics , Nucleophosmin
17.
Cancer Res ; 67(11): 5148-55, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545593

ABSTRACT

The t(2;11)(q31;p15) chromosomal translocation results in a fusion between the NUP98 and HOXD13 genes and has been observed in patients with myelodysplastic syndrome (MDS) or acute myelogenous leukemia. We previously showed that expression of the NUP98-HOXD13 (NHD13) fusion gene in transgenic mice results in an invariably fatal MDS; approximately one third of mice die due to complications of severe pancytopenia, and about two thirds progress to a fatal acute leukemia. In the present study, we used retroviral insertional mutagenesis to identify genes that might collaborate with NHD13 as the MDS transformed to an acute leukemia. Newborn NHD13 transgenic mice and littermate controls were infected with the MOL4070LTR retrovirus. The onset of leukemia was accelerated, suggesting a synergistic effect between the NHD13 transgene and the genes neighboring retroviral insertion events. We identified numerous common insertion sites located near protein-coding genes and confirmed dysregulation of a subset of these by expression analyses. Among these genes were Meis1, a known collaborator of HOX and NUP98-HOX fusion genes, and Mn1, a transcriptional coactivator involved in human leukemia through fusion with the TEL gene. Other putative collaborators included Gata2, Erg, and Epor. Of note, we identified a common insertion site that was >100 kb from the nearest coding gene, but within 20 kb of the miR29a/miR29b1 microRNA locus. Both of these miRNA were up-regulated, demonstrating that retroviral insertional mutagenesis can target miRNA loci as well as protein-coding loci. Our data provide new insights into NHD13-mediated leukemogenesis as well as retroviral insertional mutagenesis mechanisms.


Subject(s)
Homeodomain Proteins/genetics , Leukemia, Myeloid/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cloning, Molecular , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , GATA2 Transcription Factor/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Moloney murine leukemia virus/genetics , Mutagenesis, Insertional , Myeloid Ecotropic Viral Integration Site 1 Protein , NIH 3T3 Cells , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Trans-Activators , Tumor Suppressor Proteins
18.
Neoplasia ; 9(4): 315-21, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17460775

ABSTRACT

In the most common form of stem cell leukemia (SCL) gene rearrangement, an interstitial deletion of 82 kb brings SCL under the control of regulatory elements that normally govern expression of the ubiquitously expressed SCL interrupting locus (SIL) gene, which is located directly upstream of SCL. To investigate the effect of this fusion in a mouse model, a bacterial artificial chromosome (BAC) clone containing both human SIL and SCL genes was isolated, and loxP sites were inserted into intron 1 of both the SIL and SCL genes, corresponding to the sites at which recombination occurs in human T-cell acute lymphocytic leukemia patients. This BAC clone was used to generate transgenic SILloxloxSCL mice. These transgenic mice were subsequently bred to Lck-Cre mice that express the Cre recombinase specifically in the thymus. The BAC transgene was recombined between the two loxP sites in over 50% of the thymocytes from SILloxloxSCL/Cre double-transgenic mice, bringing the SCL gene under the direct control of SIL regulatory elements. Aberrant SCL gene expression in the thymus was verified by reverse transcription-polymerase chain reaction. Using FACS analysis, we found that mice carrying both SILloxloxSCL and Cre transgenes have increased CD4-/CD8- thymocytes compared with transgene-negative mice. In the spleen, these transgenic mice show a marked reduction in the number of mature CD4+ or CD8+ cells. These results demonstrate that conditional activation of SCL under control of SIL regulatory elements can impair normal T-cell development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , CD4-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/enzymology , Cell Differentiation/genetics , Extracellular Matrix Proteins/physiology , Gene Rearrangement, T-Lymphocyte/genetics , Integrases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein-Lysine 6-Oxidase/physiology , Proto-Oncogene Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Humans , Integrases/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, SCID , Mice, Transgenic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Recombination, Genetic/immunology , T-Cell Acute Lymphocytic Leukemia Protein 1
19.
Cancer Res ; 65(16): 7151-8, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103065

ABSTRACT

OLIG2 (originally designated BHLHB1) encodes a transcription factor that contains the basic helix-loop-helix motif. Although expression of OLIG2 is normally restricted to neural tissues, overexpression of OLIG2 has been shown in patients with precursor T-cell lymphoblastic lymphoma/leukemia (pre-T LBL). In the current study, we found that overexpression of OLIG2 was not only found in oligodendroglioma samples and normal neural tissue but also in a wide spectrum of malignant cell lines including leukemia, non-small cell lung carcinoma, melanoma, and breast cancer cell lines. To investigate whether enforced expression of OLIG2 is oncogenic, we generated transgenic mice that overexpressed OLIG2 in the thymus. Ectopic OLIG2 expression in the thymus was only weakly oncogenic as only 2 of 85 mice developed pre-T LBL. However, almost 60% of transgenic mice that overexpressed both OLIG2 and LMO1 developed pre-T LBL with large thymic tumor masses. Gene expression profiling of thymic tumors that developed in OLIG2/LMO1 mice revealed up-regulation of Notch1 as well as Deltex1 (Dtx1) and pre T-cell antigen receptor alpha (Ptcra), two genes that are considered to be downstream of Notch1. Of note, we found mutations in the Notch1 heterodimerization or proline-, glutamic acid-, serine-, and threonine-rich domain in three of six primary thymic tumors. In addition, growth of leukemic cell lines established from OLIG2/LMO1 transgenic mice was suppressed by a gamma-secretase inhibitor, suggesting that Notch1 up-regulation is important for the proliferation of OLIG2-LMO1 leukemic cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , Leukemia, T-Cell/genetics , Nerve Tissue Proteins/biosynthesis , Oncogene Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Endopeptidases , Gene Expression Profiling , Humans , LIM Domain Proteins , Leukemia, T-Cell/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1 , Receptors, Cell Surface/genetics , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Transcription Factors/genetics
20.
Blood ; 106(1): 287-95, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15755899

ABSTRACT

The myelodysplastic syndromes (MDSs) are a group of clonal hematopoietic stem-cell disorders characterized by ineffective hematopoiesis and dysplasia. A wide spectrum of genetic aberrations has been associated with MDS, including chromosomal translocations involving the NUP98 gene. Using a NUP98-HOXD13 fusion gene, we have developed a mouse model that faithfully recapitulates all of the key features of MDS, including peripheral blood cytopenias, bone marrow dysplasia, and apoptosis, and transformation to acute leukemia. The MDS that develops in NUP98-HOXD13 transgenic mice is uniformly fatal. Within 14 months, all of the mice died of either leukemic transformation or severe anemia and leucopenia as a result of progressive MDS. The NUP98-HOXD13 fusion gene inhibits megakaryocytic differentiation and increases apoptosis in the bone marrow, suggesting a mechanism leading to ineffective hematopoiesis in the presence of a hypercellular bone marrow. These mice provide an accurate preclinical model that can be used for the evaluation of MDS therapy and biology.


Subject(s)
Homeodomain Proteins/genetics , Leukemia/genetics , Leukemia/physiopathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Nuclear Pore Complex Proteins/genetics , Transcription Factors/genetics , Acute Disease , Animals , Carcinogens/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Transformation, Neoplastic , Disease Models, Animal , Humans , K562 Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Penetrance , Phenotype , Severity of Illness Index , Tetradecanoylphorbol Acetate/pharmacology , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...