Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38106192

ABSTRACT

Chromothripsis, the process of catastrophic shattering and haphazard repair of chromosomes, is a common event in cancer. Whether chromothripsis might constitute an actionable molecular event amenable to therapeutic targeting remains an open question. We describe recurrent chromothripsis of chromosome 21 in a subset of patients in blast phase of a myeloproliferative neoplasm (BP-MPN), which alongside other structural variants leads to amplification of a region of chromosome 21 in ∼25% of patients ('chr21amp'). We report that chr21amp BP-MPN has a particularly aggressive and treatment-resistant phenotype. The chr21amp event is highly clonal and present throughout the hematopoietic hierarchy. DYRK1A , a serine threonine kinase and transcription factor, is the only gene in the 2.7Mb minimally amplified region which showed both increased expression and chromatin accessibility compared to non-chr21amp BP-MPN controls. We demonstrate that DYRK1A is a central node at the nexus of multiple cellular functions critical for BP-MPN development, including DNA repair, STAT signalling and BCL2 overexpression. DYRK1A is essential for BP-MPN cell proliferation in vitro and in vivo , and DYRK1A inhibition synergises with BCL2 targeting to induce BP-MPN cell apoptosis. Collectively, these findings define the chr21amp event as a prognostic biomarker in BP-MPN and link chromothripsis to a druggable target.

2.
Blood Cancer J ; 7(6): e572, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28622305

ABSTRACT

Myelofibrosis (MF) is characterized by hyperactivation of thrombopoietin (TPO) signaling, which induces a RPS14 deficiency that de-regulates GATA1 in megakaryocytes by hampering its mRNA translation. As mice carrying the hypomorphic Gata1low mutation, which reduces the levels of Gata1 mRNA in megakaryocytes, develop MF, we investigated whether the TPO axis is hyperactive in this model. Gata1low mice contained two times more Tpo mRNA in liver and TPO in plasma than wild-type littermates. Furthermore, Gata1low LSKs expressed levels of Mpl mRNA (five times greater than normal) and protein (two times lower than normal) similar to those expressed by LSKs from TPO-treated wild-type mice. Gata1low marrow and spleen contained more JAK2/STAT5 than wild-type tissues, an indication that these organs were reach of TPO-responsive cells. Moreover, treatment of Gata1low mice with the JAK inhibitor ruxolitinib reduced their splenomegaly. Also in Gata1low mice activation of the TPO/MPL axis was associated with a RSP14 deficiency and a discordant microarray ribosome signature (reduced RPS24, RPS26 and SBDS expression). Finally, electron microscopy revealed that Gata1low megakaryocytes contained poorly developed endoplasmic reticulum with rare polysomes. In summary, Gata1low mice are a bona fide model of MF, which recapitulates the hyperactivation of the TPO/MPL/JAK2 axis observed in megakaryocytes from myelofibrotic patients.


Subject(s)
GATA1 Transcription Factor/metabolism , Primary Myelofibrosis/genetics , Ribosomal Proteins/genetics , Thrombopoietin/metabolism , Animals , Disease Models, Animal , Female , GATA1 Transcription Factor/genetics , Humans , Male , Mice , Primary Myelofibrosis/pathology
3.
Leukemia ; 31(11): 2458-2467, 2017 11.
Article in English | MEDLINE | ID: mdl-28270692

ABSTRACT

Imetelstat (GRN163L) is a specific telomerase inhibitor that has demonstrated clinical activity in patients with myeloproliferative neoplasms (MPN) and in patients with solid tumors. The antitumor effects were associated with the development of thrombocytopenia, one of the common side effects observed in patients treated with imetelstat. The events underlying these adverse effects are not apparent. In this report, we investigated the potential mechanisms that account for imetelstat's beneficial effects in MPN patients and the manner by which imetelstat treatment leads to a reduction in platelet numbers. Using a well-established system of ex vivo megakaryopoiesis, we demonstrated that imetelestat treatment affects normal megakaryocyte (MK) development by exclusively delaying maturation of MK precursor cells. By contrast, additional stages along MPN MK development were affected by imetelstat resulting in reduced numbers of assayable colony-forming unit MK and impaired MK maturation. In addition, treatment with imetelstat inhibited the secretion of fibrogenic growth factors by malignant but not by normal MK. Our results indicate that the delay observed in normal MK maturation may account for imetelstat-induced thrombocytopenia, while the more global effects of imetelstat on several stages along the hierarchy of MPN megakaryopoiesis may be responsible for the favorable clinical outcomes reported in MPN patients.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Megakaryocytes/drug effects , Niacinamide/analogs & derivatives , Telomerase/antagonists & inhibitors , Humans , Megakaryocytes/cytology , Niacinamide/pharmacology , Oligonucleotides , Polyploidy
4.
Leukemia ; 31(3): 712-719, 2017 03.
Article in English | MEDLINE | ID: mdl-27554164

ABSTRACT

Acute myelogenous leukemia (AML) is a high-risk hematopoietic malignancy caused by a variety of mutations, including genes encoding the cohesin complex. Recent studies have demonstrated that reduction in cohesin complex levels leads to enhanced self-renewal in hematopoietic stem and progenitors (HSPCs). We sought to delineate the molecular mechanisms by which cohesin mutations promote enhanced HSPC self-renewal as this represents a critical initial step during leukemic transformation. We verified that RNAi against the cohesin subunit Rad21 causes enhanced self-renewal of HSPCs in vitro through derepression of polycomb repressive complex 2 (PRC2) target genes, including Hoxa7 and Hoxa9. Importantly, knockdown of either Hoxa7 or Hoxa9 suppressed self-renewal, implying that both are critical downstream effectors of reduced cohesin levels. We further demonstrate that the cohesin and PRC2 complexes interact and are bound in close proximity to Hoxa7 and Hoxa9. Rad21 depletion resulted in decreased levels of H3K27me3 at the Hoxa7 and Hoxa9 promoters, consistent with Rad21 being critical to proper gene silencing by recruiting the PRC2 complex. Our data demonstrates that the cohesin complex regulates PRC2 targeting to silence Hoxa7 and Hoxa9 and negatively regulate self-renewal. Our studies identify a novel epigenetic mechanism underlying leukemogenesis in AML patients with cohesin mutations.


Subject(s)
Cell Self Renewal/genetics , Epigenetic Repression , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Aneuploidy , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cluster Analysis , DNA-Binding Proteins , Gene Deletion , Gene Expression Profiling , Histones/metabolism , Homeodomain Proteins/metabolism , Mice , Models, Biological , Multigene Family , Multiprotein Complexes/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , Protein Binding , Protein Interaction Domains and Motifs/genetics , Cohesins
5.
Blood Cancer J ; 6(9): e478, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27689744

ABSTRACT

FAXDC2 (fatty acid hydroxylase domain containing 2) is a member of the fatty acid hydroxylase superfamily. Given the important role of fatty acids in megakaryocytes, we have studied the role of this gene in the development of this lineage. Here we show that the expression of FAXDC2 is constantly elevated during megakaryocyte maturation. In contrast, FAXDC2 is significantly downregulated in acute myeloid leukemia and acute megakaryoblastic leukemia. Moreover, FAXDC2 overexpression promotes the differentiation of megakaryocytic cell lines and primary cells, whereas its knockdown disrupts their maturation. Mechanism study shows that FAXDC2 overexpression enhances extracellular signal-regulated kinase (ERK) signaling and increases RUNX1 (Runt-related transcription factor 1) expression. FAXDC2 also restores megakaryocytic differentiation in cells exposed to an ERK inhibitor or those expressing a dominant negative form of RUNX1. Finally, FAXDC2 overexpression leads to an increase in sphingolipid GM3 synthase, suggesting a potential role of FAXDC2 in lipid metabolism that increases ERK signaling and facilitates megakaryocyte differentiation. Together, these results show that FAXDC2 plays a novel role in development of megakaryocytes and its dysregulation may contribute to abnormal hematopoietic cell development in leukemia.

6.
Leukemia ; 30(9): 1816-23, 2016 09.
Article in English | MEDLINE | ID: mdl-27285583

ABSTRACT

Children with Down syndrome (DS) are at a 20-fold increased risk for acute lymphoblastic leukemia (DS-ALL). Although the etiology of this higher risk of developing leukemia remains largely unclear, the recent identification of CRLF2 (cytokine receptor like factor 2) and JAK2 mutations and study of the effect of trisomy of Hmgn1 and Dyrk1a (dual-specificity tyrosine phosphorylation-regulated kinase 1A) on B-cell development have shed significant new light on the disease process. Here we focus on the clinical features, biology and genetics of ALL in children with DS. We review the unique characteristics of DS-ALL on both the clinical and molecular levels and discuss the differences in treatments and outcomes in ALL in children with DS compared with those without DS. The identification of new biological insights is expected to pave the way for novel targeted therapies.


Subject(s)
Down Syndrome/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , B-Lymphocytes , Child , Humans , Molecular Targeted Therapy/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Treatment Outcome
7.
Oncogene ; 34(5): 537-45, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-24632603

ABSTRACT

The Aurora kinases, which include Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are serine/threonine kinases required for the control of mitosis (AURKA and AURKB) and meiosis (AURKC). Since their discovery nearly 20 years ago, Aurora kinases have been studied extensively in cell and cancer biology. Several early studies found that Aurora kinases are amplified and overexpressed at the transcript and protein level in various malignancies, including several types of leukemia. These discoveries and others provided a rationale for the development of small-molecule inhibitors of Aurora kinases as leukemia therapies. The first generation of Aurora kinase inhibitors did not fare well in clinical trials, owing to poor efficacy and high toxicity. However, the creation of second-generation, highly selective Aurora kinase inhibitors has increased the enthusiasm for targeting these proteins in leukemia. This review will describe the functions of each Aurora kinase, summarize their involvement in leukemia and discuss inhibitor development and efficacy in leukemia clinical trials.


Subject(s)
Aurora Kinase A/genetics , Aurora Kinase B/genetics , Aurora Kinase C/genetics , Leukemia/genetics , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase C/antagonists & inhibitors , Cell Cycle/genetics , Clinical Trials as Topic , Humans , Leukemia/drug therapy , Leukemia/pathology , Meiosis/genetics , Mitosis/genetics , Small Molecule Libraries/therapeutic use
8.
Cell Death Dis ; 5: e988, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24407241

ABSTRACT

GATA1 mutations are tightly associated with transient myeloproliferative disorder (TMD) and acute megakaryoblstic leukemia (AMKL) in children with Down syndrome. Numerous genes are altered in GATA-1-deficient megakaryocytes, which may contribute to the hyperproliferation and abnormal terminal differentiation of these malignant cells. In this study, we demonstrate that Pstpip2 is a GATA-1-repressed gene that controls megakaryopoiesis. Ectopic expression of PSTPIP2 impaired megakaryocytic differentiation as evidenced by a decrease of CD41 expression and reduced DNA content in K562 cells. PSTPIP2 overexpression also caused enhanced activation of Src family kinases and subsequently reduced ERK phosphorylation. Consistently, PSTPIP2 knockdown showed the opposite effect on differentiation and signaling. Moreover, the W232A mutant of PSTPIP2, defective in its interaction with PEST family phosphatases that recruit c-Src terminal kinase (CSK) to suppress Src family kinases, failed to inhibit differentiation and lost its ability to enhance Src family kinases or reduce ERK phosphorylation. In fact, the W232A mutant of PSTPIP2 promoted megakaryocyte differentiation. These observations suggest that PSTPIP2 recruiting PEST phosphatases somehow blocked CSK activity and led to enhanced activation of Src family kinases and reduced ERK phosphorylation, which ultimately repressed megakaryocyte differentiation. Supporting this idea, PSTPIP2 interacted with LYN and the expression of a dominant negative LYN (LYN DN) overwhelmed the inhibitory effect of PSTPIP2 on differentiation and ERK signaling. In addition, a constitutively active LYN (LYN CA) normalized the enhanced megakaryocyte differentiation and repressed ERK signaling in PSTPIP2 knockdown cells. Finally, we found that PSTPIP2 repressed ERK signaling, differentiation, and proliferation and verified that PSTPIP2 upregulation repressed megakaryocyte development in primary mouse bone marrow cells. Our study thus reveals a novel mechanism by which dysregulation of PSTPIP2 due to GATA-1 deficiency may contribute to abnormal megakaryocyte proliferation and differentiation in pathogenesis of related diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation , Cytoskeletal Proteins/genetics , Down-Regulation , GATA1 Transcription Factor/deficiency , GATA1 Transcription Factor/metabolism , Megakaryocytes/cytology , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , GATA1 Transcription Factor/genetics , Gene Expression Regulation , Humans , K562 Cells , Megakaryocytes/enzymology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , src-Family Kinases/genetics
9.
Leukemia ; 27(9): 1882-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23748344

ABSTRACT

The majority of patients with BCR-ABL1-negative myeloproliferative neoplasms (MPN) harbor mutations in JAK2 or MPL, which lead to constitutive activation of the JAK/STAT, PI3K and ERK signaling pathways. JAK inhibitors by themselves are inadequate in producing selective clonal suppression in MPN and are associated with hematopoietic toxicities. MK-2206 is a potent allosteric AKT inhibitor that was well tolerated, including no evidence of myelosuppression, in a phase I study of solid tumors. Herein, we show that inhibition of PI3K/AKT signaling by MK-2206 affected the growth of both JAK2V617F- or MPLW515L-expressing cells via reduced phosphorylation of AKT and inhibition of its downstream signaling molecules. Moreover, we demonstrate that MK-2206 synergizes with ruxolitinib in suppressing the growth of JAK2V617F-mutant SET2 cells. Importantly, MK-2206 suppressed colony formation from hematopoietic progenitor cells in patients with primary myelofibrosis and alleviated hepatosplenomegaly and reduced megakaryocyte burden in the bone marrows, livers and spleens of mice with MPLW515L-induced MPN. Together, these findings establish AKT as a rational therapeutic target in the MPNs.


Subject(s)
Myeloproliferative Disorders/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Female , Fusion Proteins, bcr-abl/deficiency , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Liver/metabolism , Liver/pathology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Mutation , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism
10.
Leukemia ; 27(6): 1339-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23380710

ABSTRACT

The requirement that leukemic Gata1 mutations be present in cells harboring trisomy 21 led to the discovery that overexpression of ERG drives aberrant megakaryopoiesis. Given that constitutive PI3K/AKT signaling is a frequent component of hematologic malignancies and the relationship between AKT and Notch in this lineage, we studied the crosstalk between AKT signaling and ERG in megakaryopoiesis. We discovered that constitutive AKT signaling is associated with a dramatic increase in apoptosis of WT megakaryocytes (MKs), but that overexpression of ERG blocks AKT-induced death. We further found that Gata1 mutations protect MKs from activated AKT-induced apoptosis. As a consequence, however, the enhanced signaling inhibits differentiation of Gata1 mutant, but not WT, MKs. Gata1 mutant cells that overexpress ERG with hyperactive AKT are characterized by diminished FOXO1/3a expression and an increased dependency on the c-Jun pathway similar to that seen in acute megakaryoblastic leukemia (AMKL) cell lines, acute myeloid leukemia (AML) with knockdown of FOXO3a, or AML with expression of myristoylated Akt. Additionally, we found that the AKT allosteric inhibitor MK2206 caused reduced cell viability and proliferation of AMKL cell lines. The contribution of aberrant AKT signaling during the ontogeny of Down syndrome-transient myeloproliferative disorder/AMKL indicates that AKT is a therapeutic target in this form of AML.


Subject(s)
GATA1 Transcription Factor/metabolism , Hematopoiesis , Leukemia, Megakaryoblastic, Acute/pathology , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Cell Line, Tumor , GATA1 Transcription Factor/genetics , Humans , Leukemia, Megakaryoblastic, Acute/genetics , Mutation , Phosphorylation , Protein Binding , Transcriptional Regulator ERG
11.
Blood Cancer J ; 1: e7, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-23471017

ABSTRACT

Immediately following the 2010 annual American Society of Hematology (ASH) meeting, the 5th International Post-ASH Symposium on Chronic Myelogenous Leukemia and BCR-ABL1-Negative Myeloproliferative Neoplasms (MPNs) took place on 7-8 December 2010 in Orlando, Florida, USA. During this meeting, the most recent advances in laboratory research and clinical practice, including those that were presented at the 2010 ASH meeting, were discussed among recognized authorities in the field. The current paper summarizes the proceedings of this meeting in BCR-ABL1-negative MPN. We provide a detailed overview of new mutations with putative epigenetic effects (TET oncogene family member 2 (TET2), additional sex comb-like 1 (ASXL1), isocitrate dehydrogenase (IDH) and enhancer of zeste homolog 2 (EZH2)) and an update on treatment with Janus kinase (JAK) inhibitors, pomalidomide, everolimus, interferon-α, midostaurin and cladribine. In addition, the new 'Dynamic International Prognostic Scoring System (DIPSS)-plus' prognostic model for primary myelofibrosis (PMF) and the clinical relevance of distinguishing essential thrombocythemia from prefibrotic PMF are discussed.

12.
Blood Cancer J ; 1(11): e40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22829077

ABSTRACT

Polycythemia vera (PV), essential thrombocythemia and primary myelofibrosis, are myeloproliferative neoplasms (MPNs) with distinct clinical features and are associated with the JAK2V617F mutation. To identify genomic anomalies involved in the pathogenesis of these disorders, we profiled 87 MPN patients using Affymetrix 250K single-nucleotide polymorphism (SNP) arrays. Aberrations affecting chr9 were the most frequently observed and included 9pLOH (n=16), trisomy 9 (n=6) and amplifications of 9p13.3-23.3 (n=1), 9q33.1-34.13 (n=1) and 9q34.13 (n=6). Patients with trisomy 9 were associated with elevated JAK2V617F mutant allele burden, suggesting that gain of chr9 represents an alternative mechanism for increasing JAK2V617F dosage. Gene expression profiling of patients with and without chr9 abnormalities (+9, 9pLOH), identified genes potentially involved in disease pathogenesis including JAK2, STAT5B and MAPK14. We also observed recurrent gains of 1p36.31-36.33 (n=6), 17q21.2-q21.31 (n=5) and 17q25.1-25.3 (n=5) and deletions affecting 18p11.31-11.32 (n=8). Combined SNP and gene expression analysis identified aberrations affecting components of a non-canonical PRC2 complex (EZH1, SUZ12 and JARID2) and genes comprising a 'HSC signature' (MLLT3, SMARCA2 and PBX1). We show that NFIB, which is amplified in 7/87 MPN patients and upregulated in PV CD34+ cells, protects cells from apoptosis induced by cytokine withdrawal.

13.
Leukemia ; 24(11): 1920-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20882051

ABSTRACT

Survivin is an inhibitor of apoptosis protein family member that has an essential role in cellular proliferation as a component of the chromosome passenger complex. Survivin is highly expressed in embryos and in proliferating adult tissues, but it is not expressed in most differentiated cells. During tumorigenesis, however, survivin expression is dramatically upregulated. Although many studies have shown that survivin is required for cancer cells, the extent to which survivin contributes to the initiation of tumors is unknown. Here we show that transgenic mice that overexpress survivin in hematopoietic cells are at an increased risk of hematologic tumors. In examining how survivin might contribute to tumorigenesis, we observed that hematopoietic cells engineered to overexpress survivin are less susceptible to apoptosis. We conclude that survivin may promote tumorigenesis by imparting a survival advantage to cells that acquire additional genetic lesions.


Subject(s)
Hematologic Neoplasms/genetics , Microtubule-Associated Proteins/genetics , Up-Regulation , Animals , Apoptosis/genetics , Cell Differentiation , Cell Survival , Flow Cytometry , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/physiology , Humans , Inhibitor of Apoptosis Proteins , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Spleen/immunology , Survivin , T-Lymphocytes/immunology
15.
Oncogene ; 26(47): 6803-15, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17934487

ABSTRACT

Red blood cells and megakaryocytes arise from a common precursor, the megakaryocyte-erythroid progenitor and share many regulators including the transcription factors GATA-1 and GFI-1B and signaling molecules such as JAK2 and STAT5. These lineages also share the distinction of being associated with rare, but aggressive malignancies that have very poor prognoses. In this review, we will briefly summarize features of normal development of red blood cells and megakaryocytes and also highlight events that lead to their leukemic transformation. It is clear that much more work needs to be done to improve our understanding of the unique biology of these leukemias and to pave the way for novel targeted therapeutics.


Subject(s)
Cell Transformation, Neoplastic/pathology , Erythroid Cells/cytology , Erythroid Cells/pathology , Megakaryocytes/cytology , Megakaryocytes/pathology , Animals , Cell Transformation, Neoplastic/genetics , Humans , Leukemia/genetics , Leukemia/pathology
16.
Leukemia ; 20(7): 1261-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16673016

ABSTRACT

Chromosome condensation is essential for proper segregation of duplicated sister chromatids in mitosis. Mammalian erythroid maturation is also associated with gradual nuclear condensation. However, few proteins that are directly involved in chromosome condensation during erythropoiesis have been identified. In this report, we show that MTB (more than blood), which was initially isolated in a yeast two-hybrid screen for proteins that interact with the basic helix-loop-helix (bHLH) protein stem cell leukemia (SCL), and later identified as the murine homolog of the condensin II subunit CAP-G2, participates in erythroid cell development. MTB interacts with SCL and another hematopoietic bHLH protein, E12, and is recruited to the nucleus by SCL and E12. In addition, MTB can repress SCL/E12-mediated transcriptional activation. Consistent with the model that MTB may function together with SCL/E12 heterodimer during erythroid cell development, MTB is highly expressed in the erythroid lineage and is upregulated upon erythroid differentiation. Moreover, overexpression of MTB promotes the terminal differentiation of the murine erythroleukemia erythroid cell line. Together, these findings demonstrate that the condensin II subunit MTB/mCAP-G2 plays a novel function during erythropoiesis and suggest that key hematopoietic transcription factors such as SCL and E12 may regulate the terminal differentiation of hematopoietic cells through the interaction with condensin complexes.


Subject(s)
Erythroid Cells/cytology , Hematopoiesis/physiology , Leukemia, Erythroblastic, Acute/physiopathology , Proteins/genetics , Transcription, Genetic/physiology , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , COS Cells , Cell Cycle Proteins , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Leukemia/metabolism , Leukemia/physiopathology , Leukemia, Erythroblastic, Acute/metabolism , Mice , Molecular Sequence Data , Multiprotein Complexes/genetics , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , T-Cell Acute Lymphocytic Leukemia Protein 1 , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 1 Protein , Two-Hybrid System Techniques
17.
Genes Dev ; 15(7): 839-44, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11297508

ABSTRACT

GATA-family transcription factors are critical to the development of diverse tissues. In particular, GATA-4 has been implicated in formation of the vertebrate heart. As the mouse Gata-4 knock-out is early embryonic lethal because of a defect in ventral morphogenesis, the in vivo function of this factor in heart development remains unresolved. To search for a requirement for Gata4 in heart development, we created mice harboring a single amino acid replacement in GATA-4 that impairs its physical interaction with its presumptive cardiac cofactor FOG-2. Gata4(ki/ki) mice die just after embryonic day (E) 12.5 exhibiting features in common with Fog2(-/-) embryos as well as additional semilunar cardiac valve defects and a double-outlet right ventricle. These findings establish an intrinsic requirement for GATA-4 in heart development. We also infer that GATA-4 function is dependent on interaction with FOG-2 and, very likely, an additional FOG protein for distinct aspects of heart formation.


Subject(s)
Coronary Vessel Anomalies/genetics , Coronary Vessels/embryology , DNA-Binding Proteins/physiology , Fetal Heart/growth & development , Heart Defects, Congenital/genetics , Transcription Factors/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Basic Helix-Loop-Helix Transcription Factors , Coronary Vessel Anomalies/embryology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic and Fetal Development/genetics , Erythroid-Specific DNA-Binding Factors , Fetal Heart/pathology , GATA4 Transcription Factor , Genes, Lethal , Gestational Age , Heart Defects, Congenital/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Morphogenesis/genetics , Mutagenesis, Site-Directed , Protein Conformation , Transcription Factors/biosynthesis , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Valine/chemistry , Zebrafish Proteins
18.
Genes Dev ; 14(19): 2515-25, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11018018

ABSTRACT

The commitment of multipotent cells to particular developmental pathways requires specific changes in their transcription factor complement to generate the patterns of gene expression characteristic of specialized cell types. We have studied the role of the GATA cofactor Friend of GATA (FOG) in the differentiation of avian multipotent hematopoietic progenitors. We found that multipotent cells express high levels of FOG mRNA, which were rapidly down-regulated upon their C/EBPbeta-mediated commitment to the eosinophil lineage. Expression of FOG in eosinophils led to a loss of eosinophil markers and the acquisition of a multipotent phenotype, and constitutive expression of FOG in multipotent progenitors blocked activation of eosinophil-specific gene expression by C/EBPbeta. Our results show that FOG is a repressor of the eosinophil lineage, and that C/EBP-mediated down-regulation of FOG is a critical step in eosinophil lineage commitment. Furthermore, our results indicate that maintenance of a multipotent state in hematopoiesis is achieved through cooperation between FOG and GATA-1. We present a model in which C/EBPbeta induces eosinophil differentiation by the coordinate direct activation of eosinophil-specific promoters and the removal of FOG, a promoter of multipotency as well as a repressor of eosinophil gene expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Carrier Proteins/metabolism , Eosinophils/cytology , Hematopoietic Stem Cells/cytology , Nuclear Proteins/metabolism , Animals , Avian Proteins , Cell Differentiation , Cell Lineage , Chick Embryo , DNA-Binding Proteins , Down-Regulation , Eosinophils/metabolism , Erythroid-Specific DNA-Binding Factors , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Membrane Glycoproteins/genetics , Models, Genetic , Myeloid Cells , Phenotype , Promoter Regions, Genetic , Transcription Factors
19.
Development ; 127(10): 2031-40, 2000 May.
Article in English | MEDLINE | ID: mdl-10769228

ABSTRACT

Members of the GATA family of zinc-finger transcription factors have critical roles in a variety of cell types. GATA-1, GATA-2 and GATA-3 are required for proliferation and differentiation of several hematopoietic lineages, whereas GATA-4, GATA-5 and GATA-6 activate cardiac and endoderm gene expression programs. Two GATA cofactors have recently been identified. Friend of GATA-1 (FOG-1) interacts with GATA-1 and is expressed principally in hematopoietic lineages, whereas FOG-2 is expressed predominantly in heart and brain. Although gene targeting experiments are consistent with an essential role for FOG-1 as an activator of GATA-1 function, reporter assays in transfected cells indicate that FOG-1 and FOG-2 can act as repressors. We have cloned a Xenopus laevis homologue of FOG that is structurally most similar to FOG-1, but is expressed predominantly in heart and brain, as well as the ventral blood island and adult spleen. Ectopic expression and explant assays demonstrate that FOG proteins can act as repressors in vivo, in part through interaction with the transcriptional co-repressor, C-terminal Binding Protein (CtBP). FOG may regulate the differentiation of red blood cells by modulating expression and activity of GATA-1 and GATA-2. We propose that the FOG proteins participate in the switch from progenitor proliferation to red blood cell maturation and differentiation.


Subject(s)
Erythrocytes/cytology , Repressor Proteins/metabolism , Zinc Fingers , Alcohol Oxidoreductases , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , Erythropoiesis/physiology , GATA1 Transcription Factor , GATA2 Transcription Factor , Gene Expression , Growth Substances/metabolism , Mesoderm , Mice , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Xenopus Proteins , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism
20.
Nat Genet ; 24(3): 266-70, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10700180

ABSTRACT

Haematopoietic development is regulated by nuclear protein complexes that coordinate lineage-specific patterns of gene expression. Targeted mutagenesis in embryonic stem cells and mice has revealed roles for the X-linked gene Gata1 in erythrocyte and megakaryocyte differentiation. GATA-1 is the founding member of a family of DNA-binding proteins that recognize the motif WGATAR through a conserved multifunctional domain consisting of two C4-type zinc fingers. Here we describe a family with X-linked dyserythropoietic anaemia and thrombocytopenia due to a substitution of methionine for valine at amino acid 205 of GATA-1. This highly conserved valine is necessary for interaction of the amino-terminal zinc finger of GATA-1 with its essential cofactor, FOG-1 (for friend of GATA-1; refs 9-12). We show that the V205M mutation abrogates the interaction between Gata-1 and Fog-1, inhibiting the ability of Gata-1 to rescue erythroid differentiation in an erythroid cell line deficient for Gata-1 (G1E). Our findings underscore the importance of FOG-1:Gata-1 associations in both megakaryocyte and erythroid development, and suggest that other X-linked anaemias or thrombocytopenias may be caused by defects in GATA1.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Cryptorchidism/genetics , DNA-Binding Proteins/genetics , Point Mutation , Thrombocytopenia/genetics , Transcription Factors/genetics , X Chromosome/genetics , Adult , Amino Acid Sequence , Amino Acid Substitution , Child , Consensus Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/physiology , Erythroid-Specific DNA-Binding Factors , Female , GATA1 Transcription Factor , Hematopoiesis/genetics , Humans , Infant, Newborn , Male , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Thrombocytopenia/congenital , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/physiology , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...