Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Mol Cancer Ther ; 19(4): 1018-1030, 2020 04.
Article in English | MEDLINE | ID: mdl-32024684

ABSTRACT

Natural killer (NK) cells play a pivotal role in controlling cancer. Multiple extracellular receptors and internal signaling nodes tightly regulate NK activation. Cyclin-dependent kinases of the mediator complex (CDK8 and CDK19) were described as a signaling intermediates in NK cells. Here, we report for the first time the development and use of CDK8/19 inhibitors to suppress phosphorylation of STAT1S727 in NK cells and to augment the production of the cytolytic molecules perforin and granzyme B (GZMB). Functionally, this resulted in enhanced NK-cell-mediated lysis of primary leukemia cells. Treatment with the CDK8/19 inhibitor BI-1347 increased the response rate and survival of mice bearing melanoma and breast cancer xenografts. In addition, CDK8/19 inhibition augmented the antitumoral activity of anti-PD-1 antibody and SMAC mimetic therapy, both agents that promote T-cell-mediated antitumor immunity. Treatment with the SMAC mimetic compound BI-8382 resulted in an increased number of NK cells infiltrating EMT6 tumors. Combination of the CDK8/19 inhibitor BI-1347, which augments the amount of degranulation enzymes, with the SMAC mimetic BI-8382 resulted in increased survival of mice carrying the EMT6 breast cancer model. The observed survival benefit was dependent on an intermittent treatment schedule of BI-1347, suggesting the importance of circumventing a hyporesponsive state of NK cells. These results suggest that CDK8/19 inhibitors can be combined with modulators of the adaptive immune system to inhibit the growth of solid tumors, independent of their activity on cancer cells, but rather through promoting NK-cell function.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/drug therapy , Melanoma, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation , Cytotoxicity, Immunologic/immunology , Female , Humans , Killer Cells, Natural/drug effects , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Phosphorylation , STAT1 Transcription Factor/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Nat Immunol ; 18(2): 184-195, 2017 02.
Article in English | MEDLINE | ID: mdl-27992400

ABSTRACT

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Histone Demethylases/metabolism , Natural Killer T-Cells/physiology , Animals , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Histone Demethylases/genetics , Immunity, Innate/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nat Commun ; 6: 10116, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626423

ABSTRACT

Mammalian oocytes are arrested at prophase I until puberty when hormonal signals induce the resumption of meiosis I and progression to meiosis II. Meiotic progression is controlled by CDK1 activity and is accompanied by dynamic epigenetic changes. Although the signalling pathways regulating CDK1 activity are well defined, the functional significance of epigenetic changes remains largely unknown. Here we show that LSD1, a lysine demethylase, regulates histone H3 lysine 4 di-methylation (H3K4me2) in mouse oocytes and is essential for meiotic progression. Conditional deletion of Lsd1 in growing oocytes results in precocious resumption of meiosis and spindle and chromosomal abnormalities. Consequently, most Lsd1-null oocytes fail to complete meiosis I and undergo apoptosis. Mechanistically, upregulation of CDC25B, a phosphatase that activates CDK1, is responsible for precocious meiotic resumption and also contributes to subsequent spindle and chromosomal defects. Our findings uncover a functional link between LSD1 and the major signalling pathway governing meiotic progression.


Subject(s)
Histone Demethylases/metabolism , Meiosis , Oocytes/cytology , Oocytes/enzymology , Up-Regulation , cdc25 Phosphatases/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Female , Gene Expression Regulation , Histone Demethylases/genetics , Mice , Mice, Inbred C57BL , Signal Transduction , cdc25 Phosphatases/metabolism
5.
Mol Cell Biol ; 35(24): 4199-211, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26438601

ABSTRACT

The lysine (K)-specific demethylase (LSD1) family of histone demethylases regulates chromatin structure and the transcriptional potential of genes. LSD1 is frequently deregulated in tumors, and depletion of LSD1 family members causes developmental defects. Here, we report that reductions in the expression of the Pumilio (PUM) translational repressor complex enhanced phenotypes due to dLsd1 depletion in Drosophila. We show that the PUM complex is a target of LSD1 regulation in fly and mammalian cells and that its expression is inversely correlated with LSD1 levels in human bladder carcinoma. Unexpectedly, we find that PUM posttranscriptionally regulates LSD1 family protein levels in flies and human cells, indicating the existence of feedback loops between the LSD1 family and the PUM complex. Our results highlight a new posttranscriptional mechanism regulating LSD1 activity and suggest that the feedback loop between the LSD1 family and the PUM complex may be functionally important during development and in human malignancies.


Subject(s)
Drosophila Proteins/metabolism , Feedback, Physiological , Oxidoreductases, N-Demethylating/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Drosophila , Drosophila Proteins/biosynthesis , HeLa Cells , Histone Demethylases/metabolism , Humans , MCF-7 Cells , Mice , RNA Interference , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Small Interfering , RNA-Binding Proteins/biosynthesis , Urinary Bladder Neoplasms/pathology
6.
EMBO J ; 34(6): 759-77, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25564442

ABSTRACT

Scl/Tal1 confers hemogenic competence and prevents ectopic cardiomyogenesis in embryonic endothelium by unknown mechanisms. We discovered that Scl binds to hematopoietic and cardiac enhancers that become epigenetically primed in multipotent cardiovascular mesoderm, to regulate the divergence of hematopoietic and cardiac lineages. Scl does not act as a pioneer factor but rather exploits a pre-established epigenetic landscape. As the blood lineage emerges, Scl binding and active epigenetic modifications are sustained in hematopoietic enhancers, whereas cardiac enhancers are decommissioned by removal of active epigenetic marks. Our data suggest that, rather than recruiting corepressors to enhancers, Scl prevents ectopic cardiogenesis by occupying enhancers that cardiac factors, such as Gata4 and Hand1, use for gene activation. Although hematopoietic Gata factors bind with Scl to both activated and repressed genes, they are dispensable for cardiac repression, but necessary for activating genes that enable hematopoietic stem/progenitor cell development. These results suggest that a unique subset of enhancers in lineage-specific genes that are accessible for regulators of opposing fates during the time of the fate decision provide a platform where the divergence of mutually exclusive fates is orchestrated.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/cytology , Mesoderm/embryology , Myoblasts, Cardiac/cytology , Proto-Oncogene Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression Profiling , Gene Library , Hematopoietic Stem Cells/physiology , Humans , Mesoderm/metabolism , Microarray Analysis , Models, Biological , Molecular Sequence Data , Myoblasts, Cardiac/physiology , Sequence Analysis, RNA , T-Cell Acute Lymphocytic Leukemia Protein 1
7.
Blood ; 123(20): 3175-84, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24652990

ABSTRACT

The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor ß subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.


Subject(s)
Co-Repressor Proteins/metabolism , Erythropoiesis , Animals , Cells, Cultured , Co-Repressor Proteins/genetics , Cytokine Receptor Common beta Subunit/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Myeloid Cells/cytology , Receptors, Interleukin-3/metabolism , Signal Transduction
8.
Bio Protoc ; 4(5)2014 May 03.
Article in English | MEDLINE | ID: mdl-27525296

ABSTRACT

Hematopoietic differentiation is a highly complex process originating from an extraordinary population of cells called long-term repopulating hematopoietic stem cells (LT-HSCs). The unique feature of all stem cells, including HSCs, is their exceptional ability to divide asymmetrically giving rise to two different kinds of offspring. One daughter cell becomes an LT-HSC itself (self-renews) to maintain the LT-HSC pool, whereas the second daughter cell pursues a differentiation fate to ultimately give rise to terminally differentiated mature blood cells (Orkin and Zon, 2008). Quantification of phenotypic LT-HSCs can be performed by multi-color flow cytometry and the gold standard for assessment of LT-HSC self-renewal and function is competitive bone marrow transplantation (Miller et al., 2008). Although these methods are irreplaceable to determine LT-HSC abundance and functionality, they have their disadvantages and limitations. For example, competitive bone marrow transplantation is typically monitored as a function of peripheral blood donor contribution over 12-16 weeks. While reduced peripheral blood donor contribution by itself signifies impairment in the stem/progenitor cells compartment, it cannot unambiguously discriminate between reduced LT-HSC self-renewal, impaired LT-HSC differentiation or compromised progenitor cell differentiation. Here we describe an LT-HSCs methylcellulose colony-forming assay, as a fast complementary in vitro method to directly assess LT-HSC differentiation capacity. As described in Kerenyi et al. (2013), this technique acts as a powerful tool to differentiate between LT-HSC or progenitor cell differentiation defects.

9.
Cell Stem Cell ; 13(4): 492-505, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24035353

ABSTRACT

Stem cell differentiation pathways are most often studied at the population level, whereas critical decisions are executed at the level of single cells. We have established a highly multiplexed, quantitative PCR assay to profile in an unbiased manner a panel of all commonly used cell surface markers (280 genes) from individual cells. With this method, we analyzed over 1,500 single cells throughout the mouse hematopoietic system and illustrate its utility for revealing important biological insights. The comprehensive single cell data set permits mapping of the mouse hematopoietic stem cell differentiation hierarchy by computational lineage progression analysis. Further profiling of 180 intracellular regulators enabled construction of a genetic network to assign the earliest differentiation event during hematopoietic lineage specification. Analysis of acute myeloid leukemia elicited by MLL-AF9 uncovered a distinct cellular hierarchy containing two independent self-renewing lineages with different clonal activities. The strategy has broad applicability in other cellular systems.


Subject(s)
Antigens, Surface/analysis , Antigens, Surface/genetics , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic System/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Polymerase Chain Reaction
10.
Nat Chem Biol ; 9(10): 643-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974116

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is the histone lysine N-methyltransferase component of the Polycomb repressive complex 2 (PRC2), which, in conjunction with embryonic ectoderm development (EED) and suppressor of zeste 12 homolog, regulates cell lineage determination and homeostasis. Enzymatic hyperactivity has been linked to aberrant repression of tumor suppressor genes in diverse cancers. Here, we report the development of stabilized α-helix of EZH2 (SAH-EZH2) peptides that selectively inhibit H3 Lys27 trimethylation by dose-responsively disrupting the EZH2-EED complex and reducing EZH2 protein levels, a mechanism distinct from that reported for small-molecule EZH2 inhibitors targeting the enzyme catalytic domain. MLL-AF9 leukemia cells, which are dependent on PRC2, undergo growth arrest and monocyte-macrophage differentiation upon treatment with SAH-EZH2, consistent with observed changes in expression of PRC2-regulated, lineage-specific marker genes. Thus, by dissociating the EZH2-EED complex, we pharmacologically modulate an epigenetic 'writer' and suppress PRC2-dependent cancer cell growth.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Peptides/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enhancer of Zeste Homolog 2 Protein , Humans , Leukemia/metabolism , Leukemia/pathology , Models, Molecular , Peptides/chemical synthesis , Peptides/chemistry , Polycomb Repressive Complex 2/metabolism , Structure-Activity Relationship
11.
Elife ; 2: e00633, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23795291

ABSTRACT

Here, we describe that lysine-specific demethylase 1 (Lsd1/KDM1a), which demethylates histone H3 on Lys4 or Lys9 (H3K4/K9), is an indispensible epigenetic governor of hematopoietic differentiation. Integrative genomic analysis, combining global occupancy of Lsd1, genome-wide analysis of its substrates H3K4 monomethylation and dimethylation, and gene expression profiling, reveals that Lsd1 represses hematopoietic stem and progenitor cell (HSPC) gene expression programs during hematopoietic differentiation. We found that Lsd1 acts at transcription start sites, as well as enhancer regions. Loss of Lsd1 was associated with increased H3K4me1 and H3K4me2 methylation on HSPC genes and gene derepression. Failure to fully silence HSPC genes compromised differentiation of hematopoietic stem cells as well as mature blood cell lineages. Collectively, our data indicate that Lsd1-mediated concurrent repression of enhancer and promoter activity of stem and progenitor cell genes is a pivotal epigenetic mechanism required for proper hematopoietic maturation. DOI:http://dx.doi.org/10.7554/eLife.00633.001.


Subject(s)
Hematopoietic Stem Cells/cytology , Histone Demethylases/physiology , Cell Differentiation , Gene Expression Profiling , Histone Demethylases/genetics , Humans
12.
Proc Natl Acad Sci U S A ; 110(16): 6518-23, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23576758

ABSTRACT

Reactivation of fetal hemoglobin (HbF) in adults ameliorates the severity of the common ß-globin disorders. The transcription factor BCL11A is a critical modulator of hemoglobin switching and HbF silencing, yet the molecular mechanism through which BCL11A coordinates the developmental switch is incompletely understood. Particularly, the identities of BCL11A cooperating protein complexes and their roles in HbF expression and erythroid development remain largely unknown. Here we determine the interacting partner proteins of BCL11A in erythroid cells by a proteomic screen. BCL11A is found within multiprotein complexes consisting of erythroid transcription factors, transcriptional corepressors, and chromatin-modifying enzymes. We show that the lysine-specific demethylase 1 and repressor element-1 silencing transcription factor corepressor 1 (LSD1/CoREST) histone demethylase complex interacts with BCL11A and is required for full developmental silencing of mouse embryonic ß-like globin genes and human γ-globin genes in adult erythroid cells in vivo. In addition, LSD1 is essential for normal erythroid development. Furthermore, the DNA methyltransferase 1 (DNMT1) is identified as a BCL11A-associated protein in the proteomic screen. DNMT1 is required to maintain HbF silencing in primary human adult erythroid cells. DNMT1 haploinsufficiency combined with BCL11A deficiency further enhances γ-globin expression in adult animals. Our findings provide important insights into the mechanistic roles of BCL11A in HbF silencing and clues for therapeutic targeting of BCL11A in ß-hemoglobinopathies.


Subject(s)
Carrier Proteins/pharmacology , Co-Repressor Proteins/metabolism , Fetal Hemoglobin/metabolism , Gene Expression Regulation, Developmental/physiology , Multiprotein Complexes/metabolism , Nuclear Proteins/pharmacology , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Chromatography, Liquid , Erythroid Precursor Cells , Humans , Mice , Nuclear Proteins/metabolism , Proteomics , RNA Interference , Real-Time Polymerase Chain Reaction , Repressor Proteins , Tandem Mass Spectrometry , beta-Globins/metabolism
13.
Mol Cell ; 49(5): 934-46, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23395001

ABSTRACT

To provide a lifelong supply of blood cells, hematopoietic stem cells (HSCs) need to carefully balance both self-renewing cell divisions and quiescence. Although several regulators that control this mechanism have been identified, we demonstrate that the transcription factor PU.1 acts upstream of these regulators. So far, attempts to uncover PU.1's role in HSC biology have failed because of the technical limitations of complete loss-of-function models. With the use of hypomorphic mice with decreased PU.1 levels specifically in phenotypic HSCs, we found reduced HSC long-term repopulation potential that could be rescued completely by restoring PU.1 levels. PU.1 prevented excessive HSC division and exhaustion by controlling the transcription of multiple cell-cycle regulators. Levels of PU.1 were sustained through autoregulatory PU.1 binding to an upstream enhancer that formed an active looped chromosome architecture in HSCs. These results establish that PU.1 mediates chromosome looping and functions as a master regulator of HSC proliferation.


Subject(s)
Adult Stem Cells/metabolism , Cell Cycle/genetics , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Adult Stem Cells/pathology , Animals , Cell Proliferation , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred Strains , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism
14.
EMBO Mol Med ; 4(4): 283-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22323443

ABSTRACT

The large difference in phenotypes among tumour populations may stem from the stochastic origin of tumours from distinct cells - tumour cells are assumed to retain the phenotypes of the cells from which they derive. Yet, functional studies addressing the cellular origin of leukaemia are lacking. Here we show that the cells of origin of both, BCR/ABL-induced chronic myeloid (CML) and B-cell acute lymphoid leukaemia (B-ALL), resemble long-term haematopoietic stem cells (LT-HSCs). During disease-maintenance, CML LT-HSCs persist to function as cancer stem cells (CSCs) that maintain leukaemia and require signalling by the transcription factor STAT5. In contrast, B-ALL LT-HSCs differentiate into CSCs that correspond to pro-B cells. This transition step requires a transient IL-7 signal and is lost in IL-7Rα-deficient cells. Thus, in BCR/ABLp185(+) B-ALL and BCR/ABLp210(+) CML, the final phenotype of the tumour as well as the abundance of CSCs is dictated by diverging differentiation fates of their common cells of origin.


Subject(s)
Cell Transformation, Neoplastic/pathology , Leukemia, Basophilic, Acute/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Basophilic, Acute/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/metabolism , STAT5 Transcription Factor/metabolism
15.
J Exp Med ; 207(12): 2537-41, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21098097

ABSTRACT

A relatively small cadre of lineage-restricted transcription factors largely orchestrates erythropoiesis, but how these nuclear factors interact to regulate this complex biology is still largely unknown. However, recent technological advances, such as chromatin immunoprecipitation (ChIP) paired with massively parallel sequencing (ChIP-seq), gene expression profiling, and comprehensive bioinformatic analyses, offer new insights into the intricacies of red cell molecular circuits.


Subject(s)
Erythropoiesis , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Chromatin Immunoprecipitation , Computational Biology , GATA1 Transcription Factor/physiology , Gene Expression Profiling , Humans , Kruppel-Like Transcription Factors/physiology , Proto-Oncogene Proteins/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1
16.
Blood ; 116(9): 1548-58, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20508164

ABSTRACT

Stat5 transcription factors are essential gene regulators promoting proliferation, survival, and differentiation of all hematopoietic cell types. Mutations or fusions of oncogenic tyrosine kinases often result in constitutive Stat5 activation. We have modeled persistent Stat5 activity by using an oncogenic Stat5a variant (cS5). To analyze the hitherto unrecognized role of Stat5 serine phosphorylation in this context, we have generated cS5 constructs with mutated C-terminal serines 725 and 779, either alone or in combination. Genetic complementation assays in primary Stat5(null/null) mast cells and Stat5(DeltaN) T cells demonstrated reconstitution of proliferation with these mutants. Similarly, an in vivo reconstitution experiment of transduced Stat5(null/null) fetal liver cells transplanted into irradiated wild-type recipients revealed that these mutants exhibit biologic activity in lineage differentiation. By contrast, the leukemogenic potential of cS5 in bone marrow transplants decreased dramatically in cS5 single-serine mutants or was completely absent upon loss of both serine phosphorylation sites. Our data suggest that Stat5a serine phosphorylation is a prerequisite for cS5-mediated leukemogenesis. Hence, interference with Stat5a serine phosphorylation might provide a new therapeutic option for leukemia and myeloid dysplasias without affecting major functions of Stat5 in normal hematopoiesis.


Subject(s)
Cell Transformation, Neoplastic , Hematopoiesis/physiology , Leukemia/pathology , STAT5 Transcription Factor/metabolism , Serine/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Animals , Blotting, Western , Bone Marrow Transplantation , Cell Lineage , Cell Proliferation , Cells, Cultured , Female , Fetus , Flow Cytometry , Humans , Immunoenzyme Techniques , Leukemia/genetics , Leukemia/metabolism , Liver Transplantation , Male , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Phosphorylation , Precursor Cells, B-Lymphoid/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , Serine/genetics , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/genetics
17.
Am J Pathol ; 175(6): 2416-29, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893034

ABSTRACT

Recent data suggest that the signal transducer and activator of transcription (STAT)5 contributes to differentiation and growth of mast cells. It has also been described that constitutively phosphorylated STAT5 (pSTAT5) plays a pro-oncogenic role in various myeloid neoplasms. We examined the expression of pSTAT5 in neoplastic mast cells in systemic mastocytosis and asked whether the disease-related oncoprotein KIT D816V is involved in STAT5 activation. As assessed by immunohistochemistry using the anti-pSTAT5 antibody AX1, neoplastic mast cells were found to display pSTAT5 in all SM patients examined (n = 40). Expression of pSTAT5 was also demonstrable in the KIT D816V-positive mast cell leukemia cell line HMC-1. Using various staining-protocols, pSTAT5 was found to be located in both the cytoplasmic and nuclear compartment of mast cells. To define the functional role of KIT D816V in STAT5-activation, Ba/F3 cells with doxycycline-inducible expression of KIT D816V were used. In these cells, induction of KIT D816V resulted in an increased expression of pSTAT5 without substantial increase in total STAT5. Moreover, the KIT D816V-targeting kinase-inhibitor PKC412 was found to counteract expression of pSTAT5 in HMC-1 cells as well as doxycycline-induced expression of pSTAT5 in Ba/F3 cells. Finally, a dominant negative STAT5-construct was found to inhibit growth of HMC-1 cells. Together, our data show that neoplastic mast cells express cytoplasmic and nuclear pSTAT5, that KIT D816V promotes STAT5-activation, and that STAT5-activation contributes to growth of neoplastic mast cells.


Subject(s)
Mast Cells/metabolism , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/metabolism , Proto-Oncogene Proteins c-kit/genetics , STAT5 Transcription Factor/metabolism , Adult , Aged , Blotting, Western , Cell Separation , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Mutation
18.
Blood ; 113(26): 6507-8, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19556434
19.
Blood ; 113(3): 499-500, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147790
20.
Nat Cell Biol ; 10(11): 1341-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849980

ABSTRACT

Lamina-associated polypeptide (LAP) 2alpha is a chromatin-associated protein that binds A-type lamins. Mutations in both LAP2alpha and A-type lamins are linked to human diseases called laminopathies, but the molecular mechanisms are poorly understood. The A-type lamin-LAP2alpha complex interacts with and regulates retinoblastoma protein (pRb), but the significance of this interaction in vivo is unknown. Here we address the function of the A-type lamin-LAP2alpha complex with the use of LAP2alpha-deficient mice. We show that LAP2alpha loss causes relocalization of nucleoplasmic A-type lamins to the nuclear envelope and impairs pRb function. This causes inefficient cell-cycle arrest in dense fibroblast cultures and hyperproliferation of epidermal and erythroid progenitor cells in vivo, leading to tissue hyperplasia. Our results support a disease-relevant model in which LAP2alpha defines A-type lamin localization in the nucleoplasm, which in turn affects pRb-mediated regulation of progenitor cell proliferation and differentiation in highly regenerative tissues.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/physiology , Lamin Type A/metabolism , Membrane Proteins/metabolism , Stem Cells/physiology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Epidermal Cells , Lamin Type A/deficiency , Lamin Type A/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...