Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
EMBO J ; 31(6): 1494-505, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22266796

ABSTRACT

The t(8;21) and t(16;21) that are associated with acute myeloid leukaemia disrupt two closely related genes termed Myeloid Translocation Genes 8 (MTG8) and 16 (MTG16), respectively. Many of the transcription factors that recruit Mtg16 regulate haematopoietic stem and progenitor cell functions and are required to maintain stem cell self-renewal potential. Accordingly, we found that Mtg16-null bone marrow (BM) failed in BM transplant assays. Moreover, when removed from the animal, Mtg16-deficient stem cells continued to show defects in stem cell self-renewal assays, suggesting a requirement for Mtg16 in this process. Gene expression analysis indicated that Mtg16 was required to suppress the expression of several key cell-cycle regulators including E2F2, and chromatin immunoprecipitation assays detected Mtg16 near an E2A binding site within the first intron of E2F2. BrdU incorporation assays indicated that in the absence of Mtg16 more long-term stem cells were in the S phase, even after competitive BM transplantation where normal stem and progenitor cells are present, suggesting that Mtg16 plays a role in the maintenance of stem cell quiescence.


Subject(s)
Hematopoietic Stem Cells/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Cells, Cultured , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Repressor Proteins , S Phase/genetics
2.
FASEB J ; 29(3): 786-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25398765

ABSTRACT

Notch signaling largely determines intestinal epithelial cell fate. High Notch activity drives progenitors toward absorptive enterocytes by repressing secretory differentiation programs, whereas low Notch permits secretory cell assignment. Myeloid translocation gene-related 1 (MTGR1) is a transcriptional corepressor in the myeloid translocation gene/Eight-Twenty-One family. Given that Mtgr1(-/-) mice have a dramatic reduction of intestinal epithelial secretory cells, we hypothesized that MTGR1 is a key repressor of Notch signaling. In support of this, transcriptome analysis of laser capture microdissected Mtgr1(-/-) intestinal crypts revealed Notch activation, and secretory markers Mucin2, Chromogranin A, and Growth factor-independent 1 (Gfi1) were down-regulated in Mtgr1(-/-) whole intestines and Mtgr1(-/-) enteroids. We demonstrate that MTGR1 is in a complex with Suppressor of Hairless Homolog, a key Notch effector, and represses Notch-induced Hairy/Enhancer of Split 1 activity. Moreover, pharmacologic Notch inhibition using a γ-secretase inhibitor (GSI) rescued the hyperproliferative baseline phenotype in the Mtgr1(-/-) intestine and increased production of goblet and enteroendocrine lineages in Mtgr1(-/-) mice. GSI increased Paneth cell production in wild-type mice but failed to do so in Mtgr1(-/-) mice. We determined that MTGR1 can interact with GFI1, a transcriptional corepressor required for Paneth cell differentiation, and repress GFI1 targets. Overall, the data suggest that MTGR1, a transcriptional corepressor well characterized in hematopoiesis, plays a critical role in intestinal lineage allocation.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Lineage , Epithelial Cells/cytology , Intestines/cytology , Protease Inhibitors/pharmacology , Receptors, Notch/metabolism , Repressor Proteins/physiology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flow Cytometry , Immunoenzyme Techniques , Immunoprecipitation , Intestinal Mucosa/metabolism , Intestines/drug effects , Mice , Mice, Knockout , Paneth Cells/cytology , Paneth Cells/drug effects , Paneth Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
J Clin Invest ; 123(7): 3112-23, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23921131

ABSTRACT

Histone deacetylase 3 (HDAC3) contributes to the regulation of gene expression, chromatin structure, and genomic stability. Because HDAC3 associates with oncoproteins that drive leukemia and lymphoma, we engineered a conditional deletion allele in mice to explore the physiological roles of Hdac3 in hematopoiesis. We used the Vav-Cre transgenic allele to trigger recombination, which yielded a dramatic loss of lymphoid cells, hypocellular bone marrow, and mild anemia. Phenotypic and functional analysis suggested that Hdac3 was required for the formation of the earliest lymphoid progenitor cells in the marrow, but that the marrow contained 3-5 times more multipotent progenitor cells. Hdac3(-/-) stem cells were severely compromised in competitive bone marrow transplantation. In vitro, Hdac3(-/-) stem and progenitor cells failed to proliferate, and most cells remained undifferentiated. Moreover, one-third of the Hdac3(-/-) stem and progenitor cells were in S phase 2 hours after BrdU labeling in vivo, suggesting that these cells were impaired in transit through the S phase. DNA fiber-labeling experiments indicated that Hdac3 was required for efficient DNA replication in hematopoietic stem and progenitor cells. Thus, Hdac3 is required for the passage of hematopoietic stem/progenitor cells through the S phase, for stem cell functions, and for lymphopoiesis.


Subject(s)
DNA Replication , Hematopoietic Stem Cells/enzymology , Histone Deacetylases/physiology , Animals , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Hematopoietic Stem Cells/physiology , Lymphopoiesis , Mice , Mice, Inbred C57BL , Mice, Knockout , S Phase , Spleen/pathology , Transcriptome
4.
PLoS One ; 7(12): e51205, 2012.
Article in English | MEDLINE | ID: mdl-23251453

ABSTRACT

Myeloid translocation genes (MTGs) are transcriptional corepressors originally identified in acute myelogenous leukemia that have recently been linked to epithelial malignancy with non-synonymous mutations identified in both MTG8 and MTG16 in colon, breast, and lung carcinoma in addition to functioning as negative regulators of WNT and Notch signaling. A yeast two-hybrid approach was used to discover novel MTG binding partners. This screen identified the Zinc fingers, C2H2 and BTB domain containing (ZBTB) family members ZBTB4 and ZBTB38 as MTG16 interacting proteins. ZBTB4 is downregulated in breast cancer and modulates p53 responses. Because ZBTB33 (Kaiso), like MTG16, modulates Wnt signaling at the level of TCF4, and its deletion suppresses intestinal tumorigenesis in the Apc(Min) mouse, we determined that Kaiso also interacted with MTG16 to modulate transcription. The zinc finger domains of Kaiso as well as ZBTB4 and ZBTB38 bound MTG16 and the association with Kaiso was confirmed using co-immunoprecipitation. MTG family members were required to efficiently repress both a heterologous reporter construct containing Kaiso binding sites (4×KBS) and the known Kaiso target, Matrix metalloproteinase-7 (MMP-7/Matrilysin). Moreover, chromatin immunoprecipitation studies placed MTG16 in a complex occupying the Kaiso binding site on the MMP-7 promoter. The presence of MTG16 in this complex, and its contributions to transcriptional repression both required Kaiso binding to its binding site on DNA, establishing MTG16-Kaiso binding as functionally relevant in Kaiso-dependent transcriptional repression. Examination of a large multi-stage CRC expression array dataset revealed patterns of Kaiso, MTG16, and MMP-7 expression supporting the hypothesis that loss of either Kaiso or MTG16 can de-regulate a target promoter such as that of MMP-7. These findings provide new insights into the mechanisms of transcriptional control by ZBTB family members and broaden the scope of co-repressor functions for the MTG family, suggesting coordinate regulation of transcription by Kaiso/MTG complexes in cancer.


Subject(s)
Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/physiology , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Binding Sites , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Gene Knockdown Techniques , HEK293 Cells , HT29 Cells , Humans , K562 Cells , Matrix Metalloproteinase 7/genetics , Oligonucleotide Array Sequence Analysis , Transcription Factors/genetics
5.
Mol Cell Biol ; 31(13): 2544-51, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21536648

ABSTRACT

Mtg16/Eto2 is a transcriptional corepressor that is disrupted by t(16;21) in acute myeloid leukemia. Using mice lacking Mtg16, we found that Mtg16 is a critical regulator of T-cell development. Deletion of Mtg16 led to reduced thymocyte development in vivo, and after competitive bone marrow transplantation, there was a nearly complete failure of Mtg16(-/-) cells to contribute to thymocyte development. This defect was recapitulated in vitro as Mtg16(-/-) Lineage(-)/Sca1(+)/c-Kit(+) (LSK) cells of the bone marrow or DN1 cells of the thymus failed to produce CD4(+)/CD8(+) cells in response to a Notch signal. Complementation of these defects by reexpressing Mtg16 showed that 3 highly conserved domains were somewhat dispensable for T-cell development but required the capacity of Mtg16 to suppress E2A-dependent transcriptional activation and to bind to the Notch intracellular domain. Thus, Mtg16 integrates the activities of signaling pathways and nuclear factors in the establishment of T-cell fate specification.


Subject(s)
Cell Lineage , Lymphopoiesis , Nuclear Proteins/physiology , T-Lymphocytes/physiology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Transplantation , Genetic Complementation Test , Mice , Mice, Mutant Strains , Nuclear Proteins/genetics , Protein Structure, Tertiary , Receptors, Notch/metabolism , Repressor Proteins , Transcription Factors/genetics
6.
Article in English | MEDLINE | ID: mdl-21841905

ABSTRACT

Success in academic medicine requires scientific and clinical aptitude and the ability to lead a team effectively. Although combined MD/PhD training programs invest considerably in the former, they often do not provide structured educational opportunities in leadership, especially as applied to investigative medicine. To fill a critical knowledge gap in physician-scientist training, the Vanderbilt Medical Scientist Training Program (MSTP) developed a biennial two-day workshop in investigative leadership. MSTP students worked in partnership with content experts to develop a case-based curriculum and deliver the material. In its initial three offerings in 2006, 2008, and 2010, the workshop was judged by MSTP student attendees to be highly effective. The Vanderbilt MSTP Leadership Workshop offers a blueprint for collaborative student-faculty interactions in curriculum design and a new educational modality for physician-scientist training.


Subject(s)
Education/organization & administration , Leadership , Physicians , Competency-Based Education , Education, Graduate , Humans , Professional Competence , Teaching/methods
7.
Mol Cell Biol ; 30(7): 1852-63, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123979

ABSTRACT

The Notch signaling pathway regulates gene expression programs to influence the specification of cell fate in diverse tissues. In response to ligand binding, the intracellular domain of the Notch receptor is cleaved by the gamma-secretase complex and then translocates to the nucleus. There, it binds the transcriptional repressor CSL, triggering its conversion to an activator of Notch target gene expression. The events that control this conversion are poorly understood. We show that the transcriptional corepressor, MTG16, interacts with both CSL and the intracellular domains of Notch receptors, suggesting a pivotal role in regulation of the Notch transcription complex. The Notch1 intracellular domain disrupts the MTG16-CSL interaction. Ex vivo fate specification in response to Notch signal activation is impaired in Mtg16-/- hematopoietic progenitors, and restored by MTG16 expression. An MTG16 derivative lacking the binding site for the intracellular domain of Notch1 fails to restore Notch-dependent cell fate. These data suggest that MTG16 interfaces with critical components of the Notch transcription complex to affect Notch-dependent lineage allocation in hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Nuclear Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Coculture Techniques , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , Receptor, Notch1/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL