Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cells ; 33(8): 2628-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25968920

ABSTRACT

Hhex encodes a homeodomain transcription factor that is widely expressed in hematopoietic stem and progenitor cell populations. Its enforced expression induces T-cell leukemia and we have implicated it as an important oncogene in early T-cell precursor leukemias where it is immediately downstream of an LMO2-associated protein complex. Conventional Hhex knockouts cause embryonic lethality precluding analysis of adult hematopoiesis. Thus, we induced highly efficient conditional knockout (cKO) using vav-Cre transgenic mice. Hhex cKO mice were viable and born at normal litter sizes. At steady state, we observed a defect in B-cell development that we localized to the earliest B-cell precursor, the pro-B-cell stage. Most remarkably, bone marrow transplantation using Hhex cKO donor cells revealed a more profound defect in all hematopoietic lineages. In contrast, sublethal irradiation resulted in normal myeloid cell repopulation of the bone marrow but markedly impaired repopulation of T- and B-cell compartments. We noted that Hhex cKO stem and progenitor cell populations were skewed in their distribution and showed enhanced proliferation compared to WT cells. Our results implicate Hhex in the maintenance of LT-HSCs and in lineage allocation from multipotent progenitors especially in stress hematopoiesis.


Subject(s)
Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Transcription Factors/genetics
2.
Leuk Res ; 39(1): 100-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25499232

ABSTRACT

LIM domain only-2 (LMO2) overexpression in T cells induces leukemia but the molecular mechanism remains to be elucidated. In hematopoietic stem and progenitor cells, Lmo2 is part of a protein complex comprised of class II basic helix loop helix proteins, Tal1and Lyl1. The latter transcription factors heterodimerize with E2A proteins like E47 and Heb to bind E boxes. LMO2 and TAL1 or LYL1 cooperate to induce T-ALL in mouse models, and are concordantly expressed in human T-ALL. Furthermore, LMO2 cooperates with the loss of E2A suggesting that LMO2 functions by creating a deficiency of E2A. In this study, we tested this hypothesis in Lmo2-induced T-ALL cell lines. We transduced these lines with an E47/estrogen receptor fusion construct that could be forced to homodimerize with 4-hydroxytamoxifen. We discovered that forced homodimerization induced growth arrest in 2 of the 4 lines tested. The lines sensitive to E47 homodimerization accumulated in G1 and had reduced S phase entry. We analyzed the transcriptome of a resistant and a sensitive line to discern the E47 targets responsible for the cellular effects. Our results suggest that E47 has diverse effects in T-ALL but that functional deficiency of E47 is not a universal feature of Lmo2-induced T-ALL.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/metabolism , LIM Domain Proteins/metabolism , Leukemia, T-Cell/metabolism , Protein Multimerization , Proto-Oncogene Proteins/metabolism , Transcription Factor 3/metabolism , Adaptor Proteins, Signal Transducing/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , LIM Domain Proteins/genetics , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Proto-Oncogene Proteins/genetics , Response Elements , Transcription Factor 3/genetics
3.
Exp Hematol ; 42(7): 581-93.e5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24792354

ABSTRACT

In this study, we present a remarkable clonal cell line, 32080, derived from a CD2-Lmo2- transgenic T-cell leukemia with differentiation arrest at the transition from the intermediate single positive to double positive stages of T-cell development. We observed that 32080 cells had a striking variegated pattern in CD4 expression. There was cell-to-cell variability, with some cells expressing no CD4 and others expressing high CD4. The two populations were isogenic and yet differed in their rates of apoptosis and sensitivity to glucocorticoid. We sorted the 32080 line for CD4-positive or CD4-negative cells and observed them in culture. After 1 week, both sorted populations showed variegated CD4 expression, like the parental line, showing that the two populations could interconvert. We determined that cell replication was necessary to transit from CD4(+) to CD4(-) and CD4(-) to CD4(+). Lmo2 knockdown decreased CD4 expression, while inhibition of intracellular NOTCH1 or histone deacetylase activity induced CD4 expression. Enforced expression of RUNX1 repressed CD4 expression. We analyzed the CD4 locus by Histone 3 chromatin immunoprecipitation and found silencing marks in the CD4(-) cells and activating marks in the CD4(+) population. The 32080 cell line is a striking model of intermediate single positive to double positive T-cell plasticity and invokes a novel mechanism for LMO2's oncogenic functions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CD4 Antigens/genetics , Epigenesis, Genetic , LIM Domain Proteins/genetics , Leukemia, T-Cell/genetics , Proto-Oncogene Proteins/genetics , Animals , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic
4.
Stem Cells ; 31(5): 882-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23378057

ABSTRACT

LIM domain only 2 (Lmo2) is frequently deregulated in sporadic and gene therapy-induced acute T-cell lymphoblastic leukemia (T-ALL) where its overexpression is an important initiating mutational event. In transgenic and retroviral mouse models, Lmo2 expression can be enforced in multiple hematopoietic lineages but leukemia only arises from T cells. These data suggest that Lmo2 confers clonal growth advantage in T-cell progenitors. We analyzed proliferation, differentiation, and cell death in CD2-Lmo2 transgenic thymic progenitor cells to understand the cellular effects of enforced Lmo2 expression. Most impressively, Lmo2 transgenic T-cell progenitor cells were blocked in differentiation, quiescent, and immortalized in vitro on OP9-DL1 stromal cells. These cellular effects were concordant with a transcriptional signature in Lmo2 transgenic T-cell progenitor cells that is also present in hematopoietic stem cells (HSCs) and early T-cell precursor ALL. These results are significant in light of the crucial role of Lmo2 in the maintenance of the HSC. The cellular effects and transcriptional effects have implications for LMO2-dependent leukemogenesis and the treatment of LMO2-induced T-ALL.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Hematopoietic Stem Cells/cytology , LIM Domain Proteins/biosynthesis , Leukemia, T-Cell/pathology , Precursor Cells, T-Lymphoid/cytology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation/physiology , Cell Lineage , Gene Expression , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , LIM Domain Proteins/genetics , Leukemia, T-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/pathology
5.
Blood ; 118(17): 4674-81, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21878674

ABSTRACT

Cooperation of multiple mutations is thought to be required for cancer development. In previous studies, murine myeloid leukemias induced by transducing wild-type bone marrow progenitors with a SRY sex determining region Y-box 4 (Sox4)-expressing retrovirus frequently carried proviral insertions at Sfpi1, decreasing its mRNA levels, suggesting that reduced Sfpi1 expression cooperates with Sox4 in myeloid leukemia induction. In support of this hypothesis, we show here that mice receiving Sox4 virus-infected Sfpi1(ko/+) bone marrow progenitors developed myeloid leukemia with increased penetrance and shortened latency. Interestingly, Sox4 expression further decreased Sfpi1 transcription. Ectopic SOX4 expression reduced endogenous PU.1 mRNA levels in HL60 promyelocytes, and decreased Sfpi1 mRNA levels were also observed in the spleens of leukemic and preleukemic mice receiving Sox4 virus-infected wild-type bone marrow cells. In addition, Sox4 protein bound to a critical upstream regulatory element of Sfpi1 in ChIP assays. Such cooperation probably occurs in de novo human acute myeloid leukemias, as an analysis of 285 acute myeloid leukemia patient samples found a significant negative correlation between SOX4 and PU.1 expression. Our results establish a novel cooperation between Sox4 and reduced Sfpi1 expression in myeloid leukemia development and suggest that SOX4 could be an important new therapeutic target in human acute myeloid leukemia.


Subject(s)
Haploinsufficiency/physiology , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins/genetics , SOXC Transcription Factors/physiology , Trans-Activators/genetics , Animals , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Epistasis, Genetic/physiology , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Trans-Activators/metabolism , Trans-Activators/physiology
6.
Blood ; 118(14): 3911-21, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21821710

ABSTRACT

Adult T-cell leukemia/lymphoma (ATLL) is an incurable disease where most patients succumb within the first year of diagnosis. Both standard chemotherapy regimens and mAbs directed against ATLL tumor markers do not alter this aggressive clinical course. Therapeutic development would be facilitated by the discovery of genes and pathways that drive or initiate ATLL, but so far amenable drug targets have not been forthcoming. Because the IL-2 signaling pathway plays a prominent role in ATLL pathogenesis, mutational analysis of pathway components should yield interesting results. In this study, we focused on JAK3, the nonreceptor tyrosine kinase that signals from the IL-2R, where activating mutations have been found in diverse neoplasms. We screened 36 ATLL patients and 24 ethnically matched controls and found 4 patients with mutations in JAK3. These somatic, missense mutations occurred in the N-terminal FERM (founding members: band 4.1, ezrin, radixin, and moesin) domain and induced gain of function in JAK3. Importantly, we show that these mutant JAK3s are inhibited with a specific kinase inhibitor already in human clinical testing. Our findings underscore the importance of this pathway in ATLL development and offer a therapeutic handle for this incurable cancer.


Subject(s)
Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Leukemia-Lymphoma, Adult T-Cell/genetics , Mutation, Missense , Amino Acid Sequence , Animals , Cell Line , DNA Mutational Analysis , Humans , Janus Kinase 3/antagonists & inhibitors , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/metabolism , Models, Molecular , Molecular Sequence Data , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , STAT5 Transcription Factor/metabolism , Sequence Alignment
7.
PLoS Genet ; 5(5): e1000491, 2009 May.
Article in English | MEDLINE | ID: mdl-19461887

ABSTRACT

Five X-linked severe combined immunodeficiency patients (SCID-X1) successfully treated with autologous bone marrow stem cells infected ex vivo with an IL2RG-containing retrovirus subsequently developed T-cell leukemia and four contained insertional mutations at LMO2. Genetic evidence also suggests a role for IL2RG in tumor formation, although this remains controversial. Here, we show that the genes and signaling pathways deregulated in murine leukemias with retroviral insertions at Lmo2 are similar to those deregulated in human leukemias with high LMO2 expression and are highly predictive of the leukemias induced in SCID-X1 patients. We also provide additional evidence supporting the notion that IL2RG and LMO2 cooperate in leukemia induction but are not sufficient and require additional cooperating mutations. The highly concordant nature of the genetic events giving rise to mouse and human leukemias with mutations at Lmo2 are an encouraging sign to those wanting to use mice to model human cancer and may help in designing safer methods for retroviral gene therapy.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Therapy/adverse effects , Leukemia, Experimental/etiology , Leukemia-Lymphoma, Adult T-Cell/etiology , Metalloproteins/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , DNA, Neoplasm/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Interleukin Receptor Common gamma Subunit/genetics , LIM Domain Proteins , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice , Mice, SCID , Models, Genetic , Molecular Sequence Data , Mutagenesis, Insertional , Proto-Oncogene Proteins , Retroviridae/genetics , Transplantation, Autologous , Virus Integration/genetics , X-Linked Combined Immunodeficiency Diseases/complications , X-Linked Combined Immunodeficiency Diseases/genetics
8.
J Vet Diagn Invest ; 18(4): 395-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16921882

ABSTRACT

To reduce the cost of whole herd screening for bovine viral diarrhea virus persistently infected animals, the sensitivity and specificity of an antigen-capture enzyme-linked immunosorbent assay (AC-ELISA) and a microtiter virus isolation ELISA using saline from ear notch samples or pooled serum was determined. Pooled saline from ear notch samples, assayed by AC-ELISA, gave a sensitivity and specificity of 98% and 94%, respectively, for pools containing 2 samples and 72% and 100%, respectively, for pools of 5. The sensitivity of pooled ear notch or serum samples for bovine viral diarrhea virus detection by microtiter virus isolation (sensitivity < 5%) or serum samples for detection by AC-ELISA (sensitivity < 15%) is too low to be used for whole herd screening. Pooling saline from ear notch samples from 2 animals tested by AC-ELISA, however, could provide a less expensive, reliable method for whole herd screening for bovine viral diarrhea virus.


Subject(s)
Antigens, Viral/blood , Diarrhea Viruses, Bovine Viral/immunology , Diarrhea Viruses, Bovine Viral/isolation & purification , Ear/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Skin/virology , Animals , Cattle , Enzyme-Linked Immunosorbent Assay/methods , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...