Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
bioRxiv ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38562874

ABSTRACT

Survival for metastatic breast cancer is low and thus, continued efforts to treat and prevent metastatic progression are critical. Estrogen is shown to promote aggressive phenotypes in multiple cancer models irrespective of estrogen receptor (ER) status. Similarly, UDP-Glucose 6-dehydrogenase (UGDH) a ubiquitously expressed enzyme involved in extracellular matrix precursors, as well as hormone processing increases migratory and invasive properties in cancer models. While the role of UGDH in cellular migration is defined, how it intersects with and impacts hormone signaling pathways associated with tumor progression in metastatic breast cancer has not been explored. Here we demonstrate that UGDH knockdown blunts estrogen-induced tumorigenic phenotypes (migration and colony formation) in ER+ and ER- breast cancer in vitro. Knockdown of UGDH also inhibits extravasation of ER- breast cancer ex vivo, primary tumor growth and animal survival in vivo in both ER+ and ER- breast cancer. We also use single cell RNA-sequencing to demonstrate that our findings translate to a human breast cancer clinical specimen. Our findings support the role of estrogen and UGDH in breast cancer progression provide a foundation for future studies to evaluate the role of UGDH in therapeutic resistance to improve outcomes and survival for breast cancer patients.

2.
World Neurosurg ; 155: 122-134, 2021 11.
Article in English | MEDLINE | ID: mdl-34343682

ABSTRACT

Epidural spinal cord compression (ESCC) secondary to spine metastases is one of the most devastating sequelae of primary cancer as it may lead to muscle weakness, paresthesia, pain, and paralysis. Spine metastases occur through a multistep process that can result in eventual ESCC; however, the lack of a preclinical model to effectively recapitulate each step of this metastatic cascade and the symptom burden of ESCC has limited our understanding of this disease process. In this review, we discuss animal models that best recapitulate ESCC. We start with a broad discussion of commonly used models of bone metastasis and end with a focused discussion of models used to specifically study ESCC. Orthotopic models offer the most authentic recapitulation of metastasis development; however, they rarely result in symptomatic ESCC and are challenging to replicate. Conversely, models that involve injection of tumor cells directly into the bloodstream or bone better mimic the symptoms of ESCC; however, they provide limited insight into the epithelial to mesenchymal transition and natural hematogenous spread of tumor cells. Therefore, until an ideal model is created, it is critical to select an animal model that is specifically designed to answer the scientific question of interest.


Subject(s)
Disease Models, Animal , Epidural Space/pathology , Spinal Cord Compression/pathology , Spinal Neoplasms/pathology , Spinal Neoplasms/secondary , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Humans , Spinal Cord Compression/surgery , Spinal Neoplasms/surgery
4.
PLoS One ; 15(4): e0232036, 2020.
Article in English | MEDLINE | ID: mdl-32343715

ABSTRACT

The NUP98 and NUP214 nucleoporins (NUPs) are recurrently fused to heterologous proteins in leukemia. The resulting chimeric oncoproteins retain the phenylalanine-glycine (FG) repeat motifs of the NUP moiety that mediate interaction with the nuclear export receptor Crm1. NUP fusion leukemias are characterized by HOXA gene upregulation; however, their molecular pathogenesis remains poorly understood. To investigate the role of Crm1 in mediating the leukemogenic properties of NUP chimeric proteins, we took advantage of the Sequestosome-1 (SQSTM1)-NUP214 fusion. SQSTM1-NUP214 retains only a short C-terminal portion of NUP214 which contains FG motifs that mediate interaction with Crm1. We introduced point mutations targeting these FG motifs and found that the ability of the resulting SQSTM1-NUP214FGmut protein to interact with Crm1 was reduced by more than 50% compared with SQSTM1-NUP214. Mutation of FG motifs affected transforming potential: while SQSTM1-NUP214 impaired myeloid maturation and conferred robust colony formation to transduced hematopoietic progenitors in a serial replating assay, the effect of SQSTM1-NUP214FGmut was considerably diminished. Moreover, SQSTM1-NUP214 caused myeloid leukemia in all transplanted mice, whereas none of the SQSTM1-NUP214FGmut reconstituted mice developed leukemia. These oncogenic effects coincided with the ability of SQSTM1-NUP214 and SQSTM1-NUP214FGmut to upregulate the expression of Hoxa and Meis1 genes in hematopoietic progenitors. Indeed, chromatin immunoprecipitation assays demonstrated that impaired SQSTM1-NUP214 interaction with Crm1 correlated with impaired binding of the fusion protein to Hoxa and Meis1 genes. These findings highlight the importance of Crm1 in mediating the leukemogenic properties of SQSTM1-NUP214, and suggest a conserved role of Crm1 in recruiting oncoproteins to their effector genes.


Subject(s)
Homeodomain Proteins/genetics , Karyopherins/metabolism , Leukemia/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sequestosome-1 Protein/genetics , Amino Acid Motifs , Animals , Cell Line , Gene Expression Regulation, Neoplastic , Leukemia/genetics , Leukemia/pathology , Mice , Mutagenesis, Site-Directed , Neoplasm Transplantation , Nuclear Pore Complex Proteins/chemistry , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Point Mutation , Up-Regulation , Exportin 1 Protein
5.
Sci Rep ; 9(1): 5664, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952900

ABSTRACT

HOX proteins interact with PBX and MEIS cofactors, which belong to the TALE-class of homeodomain (HD)-containing transcription factors. Although the formation of HOX-PBX complexes depends on a unique conserved HOX motif called hexapeptide (HX), the additional presence of MEIS induces a remodeling of the interaction, leading to a global dispensability of the HX motif for trimeric complex formation in the large majority of HOX proteins. In addition, it was shown that the anterior HOXB3 and central HOXA7 and HOXC8 proteins could use different alternative TALE interaction motifs, with or without the HX motif, depending on the DNA-binding site and cell context. Here we dissected the molecular interaction properties of the human posterior HOXA9 protein with its TALE cofactors, PBX1 and MEIS1. Analysis was performed on different DNA-binding sites in vitro and by doing Bimolecular Fluorescence Complementation (BiFC) in different cell lines. Notably, we observed that the HOXA9-TALE interaction relies consistently on the redundant activity of the HX motif and two paralog-specific residues of the HOXA9 HD. Together with previous work, our results show that HOX proteins interact with their generic TALE cofactors through various modalities, ranging from unique and context-independent to versatile and context-dependent TALE binding interfaces.


Subject(s)
Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Binding Sites/physiology , Cell Line , Cell Line, Tumor , DNA/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , MCF-7 Cells , Neoplasm Proteins/metabolism , Protein Binding/physiology
6.
PLoS One ; 10(6): e0129776, 2015.
Article in English | MEDLINE | ID: mdl-26075887

ABSTRACT

PICALM (Phosphatidyl Inositol Clathrin Assembly Lymphoid Myeloid protein) is a ubiquitously expressed protein that plays a role in clathrin-mediated endocytosis. PICALM also affects the internalization and trafficking of SNAREs and modulates macroautophagy. Chromosomal translocations that result in the fusion of PICALM to heterologous proteins cause leukemias, and genome-wide association studies have linked PICALM Single Nucleotide Polymorphisms (SNPs) to Alzheimer's disease. To obtain insight into the biological role of PICALM, we performed gene expression studies of PICALM-deficient and PICALM-expressing cells. Pathway analysis demonstrated that PICALM expression influences the expression of genes that encode proteins involved in cholesterol biosynthesis and lipoprotein uptake. Gas Chromatography-Mass Spectrometry (GC-MS) studies indicated that loss of PICALM increases cellular cholesterol pool size. Isotopic labeling studies revealed that loss of PICALM alters increased net scavenging of cholesterol. Flow cytometry analyses confirmed that internalization of the LDL receptor is enhanced in PICALM-deficient cells as a result of higher levels of LDLR expression. These findings suggest that PICALM is required for cellular cholesterol homeostasis and point to a novel mechanism by which PICALM alterations may contribute to disease.


Subject(s)
Cholesterol/metabolism , Homeostasis , Monomeric Clathrin Assembly Proteins/metabolism , Animals , Biosynthetic Pathways/genetics , Cell Line , Gene Expression , Gene Knockout Techniques , Humans , Mice , Monomeric Clathrin Assembly Proteins/genetics , Organ Specificity , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , Receptors, LDL/metabolism
7.
Nat Commun ; 5: 4998, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25241929

ABSTRACT

Genome-wide association studies have identified several loci associated with Alzheimer's disease (AD), including proteins involved in endocytic trafficking such as PICALM/CALM (phosphatidylinositol binding clathrin assembly protein). It is unclear how these loci may contribute to AD pathology. Here we show that CALM modulates autophagy and alters clearance of tau, a protein which is a known autophagy substrate and which is causatively linked to AD, both in vitro and in vivo. Furthermore, altered CALM expression exacerbates tau-mediated toxicity in zebrafish transgenic models. CALM influences autophagy by regulating the endocytosis of SNAREs, such as VAMP2, VAMP3 and VAMP8, which have diverse effects on different stages of the autophagy pathway, from autophagosome formation to autophagosome degradation. This study suggests that the AD genetic risk factor CALM modulates autophagy, and this may affect disease in a number of ways including modulation of tau turnover.


Subject(s)
Autophagy , Monomeric Clathrin Assembly Proteins/metabolism , tau Proteins/metabolism , Animals , Autophagy-Related Protein 12 , Cell Line , Drosophila , Endocytosis , Female , Fibroblasts/metabolism , Genome-Wide Association Study , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Phagosomes , Protein Binding , RNA, Small Interfering/metabolism , Risk Factors , Small Ubiquitin-Related Modifier Proteins/metabolism , Transfection , Vesicle-Associated Membrane Protein 2/metabolism , Zebrafish
8.
Exp Hematol ; 42(12): 1022-1030.e1, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25193880

ABSTRACT

Iron, an essential nutrient for cellular growth and proliferation, enters cells via clathrin-mediated endocytosis. The clathrin assembly lymphoid myeloid (CALM) protein plays an essential role in the cellular import of iron by clathrin-mediated endocytosis. CALM-AF10 leukemias harbor a single copy of the normal CALM gene and therefore may be more sensitive to the growth-inhibitory effect of iron restriction compared with normal hematopoietic cells. We found that CALM heterozygous (CALM(HET)) murine fibroblasts exhibit signs of iron deficiency, with increased surface transferrin receptor levels and reduced growth rates. CALM(HET) hematopoietic cells are more sensitive in vitro to iron chelators than their wild type counterparts. Iron chelation also displayed toxicity toward cultured CALM(HET)CALM-AF10 leukemia cells, and this effect was additive to that of chemotherapy. In mice transplanted with CALM(HET)CALM-AF10 leukemia, we found that dietary iron restriction reduced tumor burden in the spleen. However, dietary iron restriction, used alone or in conjunction with chemotherapy, did not increase survival of mice with CALM(HET)CALM-AF10 leukemia. In summary, although CALM heterozygosity results in iron deficiency and increased sensitivity to iron chelation in vitro, our data in mice do not suggest that iron depletion strategies would be beneficial for the therapy of CALM-AF10 leukemia patients.


Subject(s)
Iron, Dietary/administration & dosage , Iron/metabolism , Leukemia, Experimental/therapy , Oncogene Proteins, Fusion/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzoates/pharmacology , Benzoates/therapeutic use , Biological Transport , Cell Line, Tumor , Chelation Therapy , Combined Modality Therapy , Deferasirox , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Fibroblasts/metabolism , Hematopoietic Stem Cells/drug effects , Heterozygote , Humans , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron, Dietary/pharmacokinetics , Leukemia, Experimental/metabolism , Mice , Mice, Knockout , Monomeric Clathrin Assembly Proteins/deficiency , Monomeric Clathrin Assembly Proteins/genetics , Radiation Chimera , Spleen/pathology , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor Burden
9.
PLoS One ; 8(8): e72261, 2013.
Article in English | MEDLINE | ID: mdl-23977266

ABSTRACT

Leukemias harboring MLL translocations are frequent in children and adults, and respond poorly to therapies. The receptor tyrosine kinase FLT3 is highly expressed in these leukemias. In vitro studies have shown that pediatric MLL-rearranged ALL cells are sensitive to FLT3 inhibitors and clinical trials are ongoing to measure their therapeutic efficacy. We sought to determine the contribution of Flt3 in the pathogenesis of MLL-rearranged leukemias using a myeloid leukemia mouse model. Bone marrow from Flt3 null mice transduced with MLL-ENL or MLL-CBP was transplanted into host mice and Flt3 (-/-) leukemias were compared to their Flt3 wild type counterparts. Flt3 deficiency did not delay disease onset and had minimal impact on leukemia characteristics. To determine the anti-leukemic effect of FLT3 inhibition we studied the sensitivity of MLL-ENL leukemia cells to the FLT3 inhibitor PKC412 ex vivo. As previously reported for human MLL-rearranged leukemias, murine MLL-ENL leukemia cells with higher Flt3 levels were more sensitive to the cytotoxicity of PKC412. Interestingly, Flt3 deficient leukemia samples also displayed some sensitivity to PKC412. Our findings demonstrate that myeloid leukemias induced by MLL-rearranged genes are not dependent upon Flt3 signaling. They also highlight the discrepancy between the sensitivity of cells to Flt3 inhibition in vitro and the lack of contribution of Flt3 to the pathogenesis of MLL-rearranged leukemias in vivo.


Subject(s)
Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Knockout , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , fms-Like Tyrosine Kinase 3/deficiency
10.
Nutrients ; 5(8): 2836-59, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23887041

ABSTRACT

Iron is essential for normal cellular function. It participates in a wide variety of cellular processes, including cellular respiration, DNA synthesis, and macromolecule biosynthesis. Iron is required for cell growth and proliferation, and changes in intracellular iron availability can have significant effects on cell cycle regulation, cellular metabolism, and cell division. Perhaps not surprisingly then, neoplastic cells have been found to have higher iron requirements than normal, non-malignant cells. Iron depletion through chelation has been explored as a possible therapeutic intervention in a variety of cancers. Here, we will review iron homeostasis in non-malignant and malignant cells, the widespread effects of iron depletion on the cell, the various iron chelators that have been explored in the treatment of cancer, and the tumor types that have been most commonly studied in the context of iron chelation.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Iron, Dietary/therapeutic use , Neoplasms/drug therapy , Anemia, Iron-Deficiency/complications , Cell Cycle Checkpoints , Cell Division , Cell Proliferation/drug effects , Chelating Agents/chemistry , Chelating Agents/pharmacology , Homeostasis/drug effects , Humans , Neoplasms/complications
11.
Blood ; 121(23): 4758-68, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23487024

ABSTRACT

The t(10;11) chromosomal translocation gives rise to the CALM-AF10 fusion gene and is found in patients with aggressive and difficult-to-treat hematopoietic malignancies. CALM-AF10-driven leukemias are characterized by HOXA gene up-regulation and a global reduction in H3K79 methylation. DOT1L, the H3K79 methyltransferase, interacts with the octapeptide/leucine zipper domain of AF10, and this region has been shown to be necessary and sufficient for CALM-AF10-mediated transformation. However, the precise role of CALM in leukemogenesis remains unclear. Here, we show that CALM contains a nuclear export signal (NES) that mediates cytoplasmic localization of CALM-AF10 and is necessary for CALM-AF10-dependent transformation. Fusions of the CALM NES (NES(CALM)-AF10) or NES motifs from heterologous proteins (ABL1, Rev, PKIA, APC) in-frame with AF10 are sufficient to immortalize murine hematopoietic progenitors in vitro. The CALM NES is essential for CALM-AF10-dependent Hoxa gene up-regulation and aberrant H3K79 methylation, possibly by mislocalization of DOT1L. Finally, we observed that CALM-AF10 leukemia cells are selectively sensitive to inhibition of nuclear export by Leptomycin B. These findings uncover a novel mechanism of leukemogenesis mediated by the nuclear export pathway and support further investigation of the utility of nuclear export inhibitors as therapeutic agents for patients with CALM-AF10 leukemias.


Subject(s)
Active Transport, Cell Nucleus/physiology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation , Leukemia, Experimental/etiology , Monomeric Clathrin Assembly Proteins/physiology , Nuclear Export Signals/genetics , Oncogene Proteins, Fusion/metabolism , Amino Acid Sequence , Animals , Antibiotics, Antineoplastic/pharmacology , Bone Marrow Transplantation , Cells, Cultured , Fatty Acids, Unsaturated/pharmacology , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , Protein Transport , Sequence Homology, Amino Acid , Survival Rate
12.
PLoS One ; 7(8): e44252, 2012.
Article in English | MEDLINE | ID: mdl-22952941

ABSTRACT

The ubiquitously expressed phosphatidylinositol binding clathrin assembly (PICALM) protein associates with the plasma membrane, binds clathrin, and plays a role in clathrin-mediated endocytosis. Alterations of the human PICALM gene are present in aggressive hematopoietic malignancies, and genome-wide association studies have recently linked the PICALM locus to late-onset Alzheimer's disease. Inactivating and hypomorphic Picalm mutations in mice cause different degrees of severity of anemia, abnormal iron metabolism, growth retardation and shortened lifespan. To understand PICALM's function, we studied the consequences of PICALM overexpression and characterized PICALM-deficient cells derived from mutant fit1 mice. Our results identify a role for PICALM in transferrin receptor (TfR) internalization and demonstrate that the C-terminal PICALM residues are critical for its association with clathrin and for the inhibitory effect of PICALM overexpression on TfR internalization. Murine embryonic fibroblasts (MEFs) that are deficient in PICALM display several characteristics of iron deficiency (increased surface TfR expression, decreased intracellular iron levels, and reduced cellular proliferation), all of which are rescued by retroviral PICALM expression. The proliferation defect of cells that lack PICALM results, at least in part, from insufficient iron uptake, since it can be corrected by iron supplementation. Moreover, PICALM-deficient cells are particularly sensitive to iron chelation. Taken together, these data reveal that PICALM plays a critical role in iron homeostasis, and offer new perspectives into the pathogenesis of PICALM-associated diseases.


Subject(s)
Homeostasis , Iron/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , Amino Acids/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Clathrin/metabolism , Embryo, Mammalian/cytology , Endocytosis/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Homeostasis/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Iron Chelating Agents/pharmacology , Iron Deficiencies , Mice , Monomeric Clathrin Assembly Proteins/chemistry , Monomeric Clathrin Assembly Proteins/deficiency , Phenotype , Protein Binding/drug effects , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
13.
Sci Signal ; 5(233): ra51, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22810897

ABSTRACT

Chemokine signaling is critical for T cell function during homeostasis and inflammation and directs T cell polarity and migration through the activation of specific intracellular pathways. Here, we uncovered a previously uncharacterized role for the Abl family tyrosine kinases Abl and Arg in the regulation of T cell-dependent inflammatory responses and showed that the Abl family kinases were required for chemokine-induced T cell polarization and migration. Our data demonstrated that Abl and Arg were activated downstream of chemokine receptors and mediated the chemokine-induced tyrosine phosphorylation of human enhancer of filamentation 1 (HEF1), an adaptor protein that is required for the activity of the guanosine triphosphatase Rap1, which mediates cell adhesion and migration. Phosphorylation of HEF1 by Abl family kinases and activation of Rap1 were required for chemokine-induced T cell migration. Mouse T cells that lacked Abl and Arg exhibited defective homing to lymph nodes and impaired migration to sites of inflammation. These findings suggest that Abl family kinases are potential therapeutic targets for the treatment of T cell-dependent immune disorders that are characterized by chemokine-mediated inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/immunology , Chemokines/metabolism , Inflammation/immunology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Cell Adhesion/immunology , Guanosine Triphosphate/metabolism , Humans , Mice , Phosphorylation , Time-Lapse Imaging
14.
Haematologica ; 96(12): 1770-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21933859

ABSTRACT

BACKGROUND: Molecular and cellular events that resulted in leukemia development are well characterized but initial engraftment and proliferation of leukemic cells in bone marrow and early modifications of the bone marrow microenvironment induced by engrafted leukemic cells remain to be clarified. DESIGN AND METHODS: After retro-orbital injection of 1,000 leukemic cells expressing Mixed Lineage Leukemia-Eleven Nineteen Leukemia fusion protein in non-conditioned syngenic mice, kinetics of leukemic burden and alterations of femoral hematopoietic populations were followed using an in vivo confocal imaging system and flow cytometry. RESULTS: Three days after injection, 5% of leukemic cells were found in femurs. Little proliferation of engrafted leukemic cells could then be detected for more than two weeks while the number of femoral leukemic cells remained stable. Twenty days after injection, leukemic cells preferentially proliferated in femoral diaphysis where they formed clusters on the surface of blood vessels and bone. B220(+) lymphoid cells were found near these leukemic cell clusters and this association is correlated with a decreased number of femoral B220(+)IgM(+) cells. Increasing the number of injected leukemic cells or conditioning recipient mice with γ-irradiation resulted in leukemic cell development in diaphysis and knee. Competition experiments indicate that proliferation but not engraftment is a rate-limiting factor of leukemic cells spreading in diaphysis. Finally, 30 days after injection leukemia developed. CONCLUSIONS: After retro-orbital injection of 1,000 leukemic cells expressing Mixed Lineage Leukemia-Eleven Nineteen Leukemia into syngenic mice, leukemic cell burden preferentially initiates in femoral diaphysis and is preceded by changes of femoral B-lymphoid populations.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Femoral Neoplasms/metabolism , Femur/metabolism , Leukemia, Biphenotypic, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocytes/pathology , DNA-Binding Proteins/genetics , Diaphyses/metabolism , Diaphyses/pathology , Femoral Neoplasms/genetics , Femoral Neoplasms/pathology , Femur/pathology , Histone-Lysine N-Methyltransferase , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Tumor Burden/genetics
15.
Cancer Res ; 69(3): 1109-16, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155294

ABSTRACT

Important biological and pathologic properties are often conserved across species. Although several mouse leukemia models have been well established, the genes deregulated in both human and murine leukemia cells have not been studied systematically. We performed a serial analysis of gene expression in both human and murine MLL-ELL or MLL-ENL leukemia cells and identified 88 genes that seemed to be significantly deregulated in both types of leukemia cells, including 57 genes not reported previously as being deregulated in MLL-associated leukemias. These changes were validated by quantitative PCR. The most up-regulated genes include several HOX genes (e.g., HOX A5, HOXA9, and HOXA10) and MEIS1, which are the typical hallmark of MLL rearrangement leukemia. The most down-regulated genes include LTF, LCN2, MMP9, S100A8, S100A9, PADI4, TGFBI, and CYBB. Notably, the up-regulated genes are enriched in gene ontology terms, such as gene expression and transcription, whereas the down-regulated genes are enriched in signal transduction and apoptosis. We showed that the CpG islands of the down-regulated genes are hypermethylated. We also showed that seven individual microRNAs (miRNA) from the mir-17-92 cluster, which are overexpressed in human MLL rearrangement leukemias, are also consistently overexpressed in mouse MLL rearrangement leukemia cells. Nineteen possible targets of these miRNAs were identified, and two of them (i.e., APP and RASSF2) were confirmed further by luciferase reporter and mutagenesis assays. The identification and validation of consistent changes of gene expression in human and murine MLL rearrangement leukemias provide important insights into the genetic base for MLL-associated leukemogenesis.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Animals , DNA Methylation , Gene Expression Profiling , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid/pathology , Mice , MicroRNAs/genetics , Myeloid Progenitor Cells/pathology , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Polymerase Chain Reaction/methods
16.
Cancer Cell ; 12(1): 23-35, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17613434

ABSTRACT

Although PML-enforced RARA homodimerization allows PML/RARA to bind DNA independently of its coreceptor RXR, the latter was identified within the PML/RARA complex. We demonstrate that a PML/RARA mutant defective for RXR binding fails to trigger APL development in transgenic mice, although it still transforms primary hematopoietic progenitors ex vivo. RXR enhances PML/RARA binding to DNA and is required for rexinoid-induced APL differentiation. In RA-treated PML/RARA-transformed cells, the absence of RXR binding results in monocytic, rather than granulocytic, differentiation. PML/RARA enhances posttranslational modifications of RXRA, including its sumoylation, suggesting that PML-bound sumoylation enzymes target RXRA and possibly other PML/RARA-bound chromatin proteins, further contributing to deregulated transcription. Thus, unexpectedly, RXR contributes to several critical aspects of in vivo transformation.


Subject(s)
Nuclear Proteins/physiology , Oncogenes , Receptors, Retinoic Acid/physiology , Retinoid X Receptors/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Processing, Post-Translational , Retinoic Acid Receptor alpha , Retinoid X Receptors/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
17.
Blood ; 109(9): 4020-2, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17202314

ABSTRACT

HOX genes, MEIS1, and FLT3 are frequently up-regulated in human myeloid leukemias. Meis1 cooperates with Hox genes to induce leukemias in mice, hypothetically the consequence of Meis1-induced Flt3 overexpression. To test this, we compared the properties of Flt3(-/-) and Flt3(+/+) progenitors transduced with Hoxa9 or Hoxa9/Meis1. In a myeloid clonogenic assay, Meis1 greatly enhanced the proliferation of Hoxa9-expressing cells, massively up-regulating Flt3 protein. However, the transforming potential of Hoxa9/Meis1 was unaltered in Flt3(-/-) cells. All mice that received Hoxa9/Meis1-transduced progenitors succumbed to rapid acute myeloid leukemias regardless of Flt3 genotype. Flt3 expression levels in leukemic blasts did not correlate with parameters reflecting their proliferative rate or their impaired differentiation. Furthermore, analysis of c-Myb expression levels in Hoxa9/Meis1-transformed cells showed that the up-regulation of this critical downstream effector was independent of Flt3. Altogether, our findings demonstrate that Flt3 is dispensable to the oncogenic cooperation of Meis1 with Hoxa9.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Leukemic , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/biosynthesis , fms-Like Tyrosine Kinase 3/biosynthesis , Animals , Cell Differentiation/genetics , Cell Line, Transformed , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Genotype , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Neoplastic Stem Cells , Proto-Oncogene Proteins c-myb/biosynthesis , Proto-Oncogene Proteins c-myb/genetics , fms-Like Tyrosine Kinase 3/genetics
18.
Genes Chromosomes Cancer ; 45(9): 846-55, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16804919

ABSTRACT

The importance of HOXA genes in T-cell acute lymphoblastic leukemia (T-ALL) has recently been recognized. We report a novel chromosomal translocation in a T-ALL patient that maps upstream of the HOXA13 gene and downstream of the BCL11B/CTIP2 locus. Analysis of HOXA gene transcription demonstrated massive expression of HOXA13, whereas the other HOXA genes were unaffected. A genomic rearrangement of the HOXA locus associated with exclusive expression of HOXA13 was observed in a second patient. This situation resembles chromosomal translocations activating genes of the TLX/HOX11 family in T-ALLs. To compare the leukemogenic properties of HOXA13 to that of TLX proteins, cohorts of lethally irradiated mice were transplanted with bone marrow transduced with a retroviral vector expressing TLX3 or HOXA13. Cells transduced with TLX3 or HOXA13 could not be detected in the peripheral blood of mice post-transplantation and none of the mice developed malignancies. Cotransduction of the HOX cofactor MEIS1 with TLX3 or HOXA13 did not alter this outcome. However, in a myeloid clonogenic assay HOXA13 and TLX3 extended the proliferation of progenitors similarly to what was observed for TLX1. Altogether, our results strongly suggest the absolute requirement for cooperative events in association with homeobox gene up-regulation to induce T-cell leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Animals , Child , Homeodomain Proteins/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Oncogene Proteins/physiology , Proto-Oncogene Proteins/physiology
19.
Oncogene ; 21(24): 3855-63, 2002 May 30.
Article in English | MEDLINE | ID: mdl-12032823

ABSTRACT

To examine the effects of Notch signaling on hematopoiesis, we transplanted mice with progenitors transduced with a constitutively active form of Notch1 (Notch1IC) or the Notch1 target genes Hes. Notch1IC-transduced cells induce T cell tumors and cannot generate B lymphocytes in vivo. Hes-transplanted mice remained healthy but cells transduced with Hes1 or Hes5 were partially impaired in their ability to differentiate into B cells. Both Hes1 and Hes5 were upregulated in the BM of Notch1IC mice and their ability to interfere with the transcriptional activity of E2A in a reporter assay was comparable to that of Notch1IC. This suggests that the inhibition of B cell development in the Notch1IC-transduced cells could be mediated by the interference of HES1/HES5 proteins with E2A. Hes1-, Hes5- and Notch1IC-transduced bone marrow cells cultured ex vivo in a colony forming assay in the presence of cytokines that promote myeloid differentiation remained very immature, indicating that the myeloid potential of these bone marrow cells was altered. Thymocytes overexpressing Hes1, Hes5 or Notch1IC matured normally into CD4 and CD8 single positive cells in vivo. Altogether our data suggest that Notch1IC induces T cell tumors independently of Hes genes but that its interference with lymphoid B and myeloid maturation is partly mediated by Hes1 and Hes5. DOI:


Subject(s)
Hematopoiesis/physiology , Homeodomain Proteins/biosynthesis , Lymphocytes/metabolism , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Receptors, Cell Surface , Animals , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors , Blotting, Northern , Bone Marrow Cells/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins/biosynthesis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Receptor, Notch1 , Receptors, Notch , Repressor Proteins/biosynthesis , Retroviridae , T-Lymphocytes/metabolism , Transcription Factor HES-1 , Transcription Factors/metabolism , Transcription, Genetic , Up-Regulation
20.
J Immunol ; 168(4): 1738-45, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11823505

ABSTRACT

To study the effects of Notch on hemopoiesis we used a bone marrow transduction/transplantation model and compared the transduced and nontransduced populations in reconstituted mice. While cells expressing a constitutively active form of murine Notch1 (Notch1IC) completely lacked B cells, a profound suppression of the B lineage was also seen in the nontransduced compartment. Experiments performed with retroviral supernatants of varying titers showed that the perturbations of B cell development among the nontransduced population correlated with the percentage of Notch1IC-transduced cells inoculated into the mice. The myeloid lineage of the Notch1IC-transplanted mice was altered as well, and this also affected the nontransduced population that had features of excessive maturation. To explore the basis of these non-cell-autonomous modifications we prepared conditioned medium from ex vivo cultures of Notch1IC-transplanted mice bone marrow and showed that it inhibited B cell maturation and promoted myeloid differentiation in a dose-dependent manner. Finally, we found that the T cell leukemia/lymphomas that occur in Notch1IC-transplanted mice were accompanied by abnormal maturation of nontransduced T cells in the bone marrow. These findings indicate that modifications of neighboring cells through non-cell-autonomous modifications take part in multiple facets of the activity of Notch on hemopoiesis.


Subject(s)
Hematopoiesis , Membrane Proteins/physiology , Receptors, Cell Surface , Transcription Factors , Animals , B-Lymphocytes/immunology , Bone Marrow Transplantation , Cell Division , Cell Lineage , Coculture Techniques , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Flow Cytometry , Immunophenotyping , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation , Myeloid Progenitor Cells/immunology , Receptor, Notch1 , T-Lymphocytes/immunology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...