Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Exp Hematol ; 121: 30-37, 2023 05.
Article in English | MEDLINE | ID: mdl-36863479

ABSTRACT

Emperipolesis between neutrophils and megakaryocytes was first identified by transmission electron microscopy. Although rare under steady-state conditions, its frequency greatly increases in myelofibrosis, the most severe of myeloproliferative neoplasms, in which it is believed to contribute to increasing the transforming growth factor (TGF)-ß microenvironmental bioavailability responsible for fibrosis. To date, the challenge of performing studies by transmission electron microscopy has hampered the study of factors that drive the pathological emperipolesis observed in myelofibrosis. We established a user-friendly confocal microscopy method that detects emperipolesis by staining with CD42b, specifically expressed on megakaryocytes, coupled with antibodies that recognize the neutrophils (Ly6b or neutrophil elastase antibody). With such an approach, we first confirmed that the bone marrow from patients with myelofibrosis and from Gata1low mice, a model of myelofibrosis, contains great numbers of neutrophils and megakaryocytes in emperipolesis. Both in patients and Gata1low mice, the emperipolesed megakaryocytes were surrounded by high numbers of neutrophils, suggesting that neutrophil chemotaxis precedes the actual emperipolesis event. Because neutrophil chemotaxis is driven by CXCL1, the murine equivalent of human interleukin 8 that is expressed at high levels by malignant megakaryocytes, we tested the hypothesis that neutrophil/megakaryocyte emperipolesis could be reduced by reparixin, an inhibitor of CXCR1/CXCR2. Indeed, the treatment greatly reduced both neutrophil chemotaxis and their emperipolesis with the megakaryocytes in treated mice. Because treatment with reparixin was previously reported to reduce both TGF-ß content and marrow fibrosis, these results identify neutrophil/megakaryocyte emperipolesis as the cellular interaction that links interleukin 8 to TGF-ß abnormalities in the pathobiology of marrow fibrosis.


Subject(s)
Emperipolesis , GATA1 Transcription Factor , Megakaryocytes , Primary Myelofibrosis , Animals , Humans , Mice , Emperipolesis/drug effects , GATA1 Transcription Factor/antagonists & inhibitors , Interleukin-8 , Megakaryocytes/metabolism , Neutrophils/metabolism , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/metabolism , Transforming Growth Factor beta/metabolism
2.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884493

ABSTRACT

CCAAT/enhancer binding protein epsilon (C/EBPε) is required for eosinophil differentiation, lineage-specific gene transcription, and expression of C/EBPε32 and shorter 27kD and 14kD isoforms is developmentally regulated during this process. We previously defined the 27kD isoform (C/EBPε27) as an antagonist of GATA-1 transactivation of the eosinophil's major basic protein-1 (MBP1) P2-promoter, showing C/EBPε27 and GATA-1 physically interact. In the current study, we used a Tat-C/EBPε27 fusion protein for cell/nuclear transduction of an eosinophil myelocyte cell line to demonstrate that C/EBPε27 is a potent repressor of MBP1 transcription. We performed structure-function analyses of C/EBPε27 mapping its repressor domains, comparing it to C/EBPε32 and C/EBPε14, using GATA-1 co-transactivation of the MBP1-P2 promoter. Results show C/EBPε27 repression of GATA-1 is mediated by its unique 68aa N-terminus combined with previously identified RDI domain. This repressor activity does not require, but is enhanced by, DNA binding via the basic region of C/EBPε27 but independent of sumoylation of the RDI core "VKEEP" sumoylation site. These findings identify the N-terminus of C/EBPε27 as the minimum repressor domain required for antagonism of GATA-1 in the eosinophil. C/EBPε27 repression of GATA-1 occurs via a combination of both C/EBPε27-GATA-1 protein-protein interaction and C/EBPε27 binding to a C/EBP site in the MBP1 promoter. The C/EBPε27 isoform may serve to titrate and/or turn off eosinophil granule protein genes like MBP1 during eosinophil differentiation, as these genes are ultimately silenced in the mature cell. Understanding the functionality of C/EBPε27 in eosinophil development may prove promising in developing therapeutics that reduce eosinophil proliferation in allergic diseases.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Eosinophils/pathology , GATA1 Transcription Factor/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/pathology , Myelin Basic Protein/metabolism , Sumoylation , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation , Cell Nucleus , Eosinophils/immunology , Eosinophils/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Myelin Basic Protein/genetics , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Tumor Cells, Cultured
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1495-1504, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34637502

ABSTRACT

Resveratrol (RSV) could promote osteogenic activity, but its clinical application has been hampered in view of its poor bioavailability. Therefore, it is desirable to identify with certainty the molecular target of its bone mass boosting function, which is crucial to the design of an effective therapeutic strategy for the optimal treatment of osteoporosis. Emerging evidence has indicated that GATA-1, an important transcription factor in megakaryocyte and erythrocyte differentiation, can directly activate autophagy in erythrocytes, alluding to its impact on bone metabolism. In light of this, we sought to determine whether GATA-1 would be a putative target by which RSV would act on osteoblast proliferation and, if so, to explore the underlying mechanism involved in the process. We examined the cell viability, colony formation, cell cyclin expression, autophagy level, and the expression levels of GATA-1 and adenosine 5'-monophosphate (AMP)-activated protein kinase α (AMPKα) in osteoblastic cell strain MC3T3-E1. The results showed that RSV promoted the proliferation process in MC3T3-E1 coupled with increased expression of GATA-1 and phosphorylated AMPKα and activated autophagy. When GATA-1 was interfered with siRNA, both autophagy and proliferation were decreased. Administration of the agonist of phosphorylated AMPKα1 (Thr172) promoted the translocation of GATA-1 into the nucleus. Based on the above results, we concluded that RSV induces the proliferation of MC3T3-E1 by increasing GATA-1 expression, which thence activates autophagy; and of note, AMPKα is one of the upstream regulators of GATA-1.


Subject(s)
AMP-Activated Protein Kinases/genetics , Autophagy/drug effects , GATA1 Transcription Factor/genetics , Osteoblasts/drug effects , Osteogenesis/drug effects , Resveratrol/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival , Cyclin D1/genetics , Cyclin D1/metabolism , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Phosphorylation , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
4.
Acta Pharmacol Sin ; 42(3): 414-421, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32555443

ABSTRACT

In the past two decades, purinergic signaling has emerged as a key regulator of hematopoiesis in physiological and pathological conditions. ADP receptor P2y12 is a crucial component of this signaling, but whether it is involved in primitive hematopoiesis remains unknown. To elucidate the function of P2y12 and provide new insights for drug development, we established a zebrafish P2y12 mutant by CRISPR/Cas 9-based genetic modification system, and investigated whether P2y12 acted as an important regulator for primitive hematopoiesis. By using mass spectrometry (MS) combined with RNA sequencing, we showed that absence of P2y12 induced excessive erythropoiesis, evidenced by significantly increased expression of mature erythrocytes marker α-globin (Hbae1 and Hbae3), ß-globin (Hbbe1 and Hbbe3). Expression pattern analysis showed that P2y12 was mainly expressed in red blood cells and endothelial cells of early zebrafish embryos. Further studies revealed that primitive erythroid progenitor marker Gata1 was markedly up-regulated. Remarkably, inhibition of Gata1 by injection of Gata1 morpholino could rescue the erythroid abnormality in P2y12 mutants. The present study demonstrates the essential role of purinergic signaling in differentiation of proerythrocytes during primitive hematopoiesis, and provides potential targets for treatment of blood-related disease and drug development.


Subject(s)
GATA1 Transcription Factor/antagonists & inhibitors , Hematopoiesis/physiology , Receptors, Purinergic P2Y12/physiology , Zebrafish Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Cell Differentiation/physiology , Embryo, Nonmammalian/physiology , Endothelium, Vascular/physiology , Erythrocytes/physiology , Female , GATA1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/physiology , Gene Knockout Techniques , Hematopoiesis/genetics , Male , Mutation , Receptors, Purinergic P2Y12/genetics , Up-Regulation/physiology , Zebrafish , Zebrafish Proteins/metabolism
5.
Cell Death Dis ; 10(6): 388, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101894

ABSTRACT

CUL4A and CUL4B are closely related members in Cullin family and can each assemble a Cullin-RING E3 ligase complex (Cullin-RING Ligase 4A or 4B, CRL4A, or CRL4B) and participate in a variety of biological processes. Previously we showed that zebrafish cul4a, but not cul4b, is essential for cardiac and pectoral fin development. Here, we have identified cul4a as a crucial regulator of primitive erythropoiesis in zebrafish embryonic development. Depletion of cul4a resulted in a striking reduction of erythroid cells due to the inhibition of erythroid differentiation. Transcript levels for early hematopoietic regulatory genes including scl, lmo2, and gata1 are significantly reduced in cul4a-deficient embryos. Mechanistically, we demonstrated that scl and gata1, the central regulators of primitive hematopoiesis for erythroid determination, are transcriptionally upregulated by cul4a. These findings demonstrate an important role for cul4a in primitive erythropoiesis and may bear implications in regeneration medicine of anemia and related diseases.


Subject(s)
Cullin Proteins/metabolism , Erythropoiesis , GATA1 Transcription Factor/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/metabolism , Cell Differentiation , Cullin Proteins/genetics , Embryo, Nonmammalian/metabolism , Embryonic Development , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Morpholinos/metabolism , Promoter Regions, Genetic , RNA Interference , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
6.
Biomed Pharmacother ; 96: 1094-1102, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29174036

ABSTRACT

Erythropoietin (EPO) produces cytoprotection in many tissues by activating the EPO receptor (EPOR); these effects include anti-oxidative stress and the inhibition of apoptosis in renal tubular cells. Moreover, EPO is clinically used in diabetic patients who suffer from chronic renal disease. However, the effect of hyperglycemia on renal EPOR expression remains unknown. Therefore, we determined the changes in renal EPOR expression in diabetic rats and investigated the role of potential factors using cultured cells. Streptozotocin-induced hyperglycemic rats (STZ rats) treated with insulin or phloridzin to correct hyperglycemia were used to investigate treated with insulin or phloridzin to correct hyperglycemia were used to investigate the EPOR changes. Potential factors, including the transducer and activator of transcription 3 (STAT3) and GATA binding protein 1 (GATA1), were identified in cultured NRK-49F cells after incubation with high glucose (HG) levels to mimic diabetic animals. Renal EPOR expression was reduced by insulin and phloridzin in STZ rats, and hyperglycemia recovered. An increase in EPORs was also reproduced in hyperglycemia-exposed NRK-49F cells and HK-2 cells, which showed a higher expression of STAT3 or GATA1. Furthermore, the application of siRNA specific to STAT3 or GATA1 attenuated the higher expression of EPORs in HG-incubated NRK-49F cells. Moreover, stattic administered at a dose that was sufficient to inhibit STAT3 restored the level of renal EPORs in diabetic rats. Taken together, the expression of renal EPORs is increased by hyperglycemia via the STAT3/GATA1 pathway and has been characterized in both diabetic rats and cultured cells.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , GATA1 Transcription Factor/metabolism , Hyperglycemia/metabolism , Kidney/metabolism , Receptors, Erythropoietin/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cattle , GATA1 Transcription Factor/antagonists & inhibitors , Humans , Kidney/drug effects , Male , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/physiology
7.
Sci Rep ; 7(1): 2551, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566697

ABSTRACT

Interferon regulatory factor 3 (IRF-3) is widely known for its prompt response against viral infection by activating the interferon system. We previously reported that E2F1, Sp1 and Sp3 regulated transcriptional activity of IRF-3. Recently, different expression patterns of IRF-3 were found in lung cancer, leading to the alternation of the immunomodulatory function in tumorigenesis. However, the mechanism of transcriptional regulation of IRF-3 in lung cancer has not been extensively studied. Here, we investigated the characterization of IRF-3 promoter and found that GATA-1 bound to a specific domain of IRF-3 promoter in vitro and in vivo. We found elevated IRF-3 and decreased GATA-1 gene expression in lung adenocarcinoma in Oncomine database. Additionally, higher IRF-3 gene expression was observed in human lung adenocarcinoma, accompanied by aberrant GATA-1 protein expression. We further analyzed the relationship of GATA-1 and IRF-3 expression in lung adenocarcinoma cell lines and found that inhibition of GATA-1 by siRNA increased the promoter activity, mRNA and protein levels of IRF-3, while over-expression of GATA-1 down-regulated IRF-3 gene expression. Taken together, we conclude that reduced GATA-1 could be responsible for the upregulation of IRF-3 in lung adenocarcinoma cells through binding with a specific domain of IRF-3 promoter.


Subject(s)
Adenocarcinoma of Lung/genetics , Carcinogenesis/genetics , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factor-3/genetics , A549 Cells , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Databases, Genetic , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/metabolism , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/metabolism , Luciferases/genetics , Luciferases/metabolism , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
8.
Development ; 143(22): 4167-4172, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27707797

ABSTRACT

Many animal embryos use nuclear ß-catenin (nß-catenin) during the segregation of endomesoderm (or endoderm) from ectoderm. This mechanism is thus likely to be evolutionarily ancient. In the ascidian embryo, nß-catenin reiteratively drives binary fate decisions between ectoderm and endomesoderm at the 16-cell stage, and then between endoderm and margin (mesoderm and caudal neural) at the 32-cell stage. At the 16-cell stage, nß-catenin activates endomesoderm genes in the vegetal hemisphere. At the same time, nß-catenin suppresses the DNA-binding activity of a maternal transcription factor, Gata.a, through a physical interaction, and Gata.a thereby activates its target genes only in the ectodermal lineage. In the present study, we found that this antagonism between nß-catenin and Gata.a also operates during the binary fate switch at the 32-cell stage. Namely, in marginal cells where nß-catenin is absent, Gata.a directly activates its target, Zic-r.b (ZicL), to specify the marginal cell lineages. Thus, the antagonistic action between nß-catenin and Gata.a is involved in two consecutive stages of germ layer segregation in ascidian embryos.


Subject(s)
Body Patterning/genetics , Ciona intestinalis/embryology , GATA1 Transcription Factor/antagonists & inhibitors , Germ Layers/embryology , beta Catenin/antagonists & inhibitors , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Ciona intestinalis/genetics , Embryo, Nonmammalian , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/metabolism , Urochordata/embryology , Urochordata/genetics , beta Catenin/genetics
9.
Blood Rev ; 30(1): 49-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26228843

ABSTRACT

Acute megakaryocytic leukemia (AMegL) is a biologically heterogenous subtype of acute myeloid leukemia (AML) that arises from megakaryocytes. Improvements in the accuracy of diagnosing AMegL as well as interest in the molecular analysis of leukemias have led to an increased amount of data available on this rare AML subtype. In this review, we will analyze the diverse molecular features unique to AMegL and how they have influenced the development of novel treatment strategies, including polyploidization. The review will also consider the data available on clinical outcomes in AMegL and how it is a poor individual prognostic factor for AML. Finally, the role of allogeneic hematopoietic stem cell transplant in AMegL will be explored.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosome Aberrations , Chromosomes, Human, Pair 3 , Hematopoietic Stem Cell Transplantation , Leukemia, Megakaryoblastic, Acute/therapy , Protein Kinase Inhibitors/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Azepines/therapeutic use , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/mortality , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocytes/drug effects , Megakaryocytes/enzymology , Megakaryocytes/pathology , Prognosis , Pyrimidines/therapeutic use , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Valproic Acid/therapeutic use
10.
BMC Bioinformatics ; 16: 349, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26511446

ABSTRACT

BACKGROUND: ChIP-seq experiments are widely used to detect and study DNA-protein interactions, such as transcription factor binding and chromatin modifications. However, downstream analysis of ChIP-seq data is currently restricted to the evaluation of signal intensity and the detection of enriched regions (peaks) in the genome. Other features of peak shape are almost always neglected, despite the remarkable differences shown by ChIP-seq for different proteins, as well as by distinct regions in a single experiment. RESULTS: We hypothesize that statistically significant differences in peak shape might have a functional role and a biological meaning. Thus, we design five indices able to summarize peak shapes and we employ multivariate clustering techniques to divide peaks into groups according to both their complexity and the intensity of their coverage function. In addition, our novel analysis pipeline employs a range of statistical and bioinformatics techniques to relate the obtained peak shapes to several independent genomic datasets, including other genome-wide protein-DNA maps and gene expression experiments. To clarify the meaning of peak shape, we apply our methodology to the study of the erythroid transcription factor GATA-1 in K562 cell line and in megakaryocytes. CONCLUSIONS: Our study demonstrates that ChIP-seq profiles include information regarding the binding of other proteins beside the one used for precipitation. In particular, peak shape provides new insights into cooperative transcriptional regulation and is correlated to gene expression.


Subject(s)
Computational Biology , Chromatin Immunoprecipitation , Cluster Analysis , DNA/chemistry , DNA/metabolism , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Knockdown Techniques , Humans , K562 Cells , Megakaryocytes/cytology , Megakaryocytes/metabolism , Protein Binding , Sequence Analysis, DNA
11.
Mol Cell Biol ; 31(13): 2632-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21555453

ABSTRACT

Recent data demonstrate that small synthetic compounds specifically targeting bromodomain proteins can modulate the expression of cancer-related or inflammatory genes. Although these studies have focused on the ability of bromodomains to recognize acetylated histones, it is increasingly becoming clear that histone-like modifications exist on other important proteins, such as transcription factors. However, our understanding of the molecular mechanisms through which these modifications modulate protein function is far from complete. The transcription factor GATA1 can be acetylated at lysine residues adjacent to the zinc finger domains, and this acetylation is essential for the normal chromatin occupancy of GATA1. We have recently identified the bromodomain-containing protein Brd3 as a cofactor that interacts with acetylated GATA1 and shown that this interaction is essential for the targeting of GATA1 to chromatin. Here we describe the structural basis for this interaction. Our data reveal for the first time the molecular details of an interaction between a transcription factor bearing multiple acetylation modifications and its cognate recognition module. We also show that this interaction can be inhibited by an acetyllysine mimic, highlighting the importance of further increasing the specificity of compounds that target bromodomain and extraterminal (BET) bromodomains in order to fully realize their therapeutic potential.


Subject(s)
GATA1 Transcription Factor/metabolism , RNA-Binding Proteins/metabolism , Acetylation , Amino Acid Sequence , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Molecular Sequence Data , Protein Structure, Secondary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Transcription Factors
12.
Circulation ; 123(15): 1633-41, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21464046

ABSTRACT

BACKGROUND: Recent studies suggest that the hematopoietic and cardiac lineages have close ontogenic origins, and that an early mesodermal cell population has the potential to differentiate into both lineages. Studies also suggest that specification of these lineages is inversely regulated. However, the transcriptional networks that govern the cell fate specification of these progenitors are incompletely defined. METHODS AND RESULTS: Here, we show that Nkx2-5 regulates the hematopoietic/erythroid fate of the mesoderm precursors early during cardiac morphogenesis. Using transgenic technologies to isolate Nkx2-5 expressing cells, we observed an induction of the erythroid molecular program, including Gata1, in the Nkx2-5-null embryos. We further observed that overexpression of Nkx2-5 with an Nkx2-5-inducible embryonic stem cell system significantly repressed Gata1 gene expression and suppressed the hematopoietic/erythroid potential, but not the endothelial potential, of the embryonic stem cells. This suppression was cell-autonomous, and was partially rescued by overexpressing Gata1. In addition, we demonstrated that Nkx2-5 binds to the Gata1 gene enhancer and represses the transcriptional activity of the Gata1 gene. CONCLUSIONS: Our results demonstrate that the hematopoietic/erythroid cell fate is suppressed via Nkx2-5 during mesodermal fate determination, and that the Gata1 gene is one of the targets that are suppressed by Nkx2-5.


Subject(s)
Down-Regulation/genetics , Embryonic Development/physiology , Embryonic Stem Cells/physiology , GATA1 Transcription Factor/antagonists & inhibitors , Gene Expression Regulation, Developmental , Heart/embryology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , GATA1 Transcription Factor/biosynthesis , GATA1 Transcription Factor/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , K562 Cells , Mice , Mice, Transgenic , Myocytes, Cardiac/physiology , Transcription Factors/genetics
13.
Blood ; 115(22): 4367-76, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20339093

ABSTRACT

Previously, we have shown that overexpression of an activated mutant of signal transducer and activator of transcription-5 (STAT5) induces erythropoiesis, impaired myelopoiesis, and an increase in long-term proliferation of human hematopoietic stem/progenitor cells. Because GATA1 is a key transcription factor involved in erythropoiesis, the involvement of GATA1 in STAT5-induced phenotypes was studied by shRNA-mediated knockdown of GATA1. CD34(+) cord blood cells were double transduced with a conditionally active STAT5 mutant and a lentiviral vector expressing a short hairpin against GATA1. Erythropoiesis was completely abolished in the absence of GATA1, indicating that STAT5-induced erythropoiesis is GATA1-dependent. Furthermore, the impaired myelopoiesis in STAT5-transduced cells was restored by GATA1 knockdown. Interestingly, early cobblestone formation was only modestly affected, and long-term growth of STAT5-positive cells was increased in the absence of GATA1, whereby high progenitor numbers were maintained. Thus, GATA1 down-regulation allowed the dissection of STAT5-induced differentiation phenotypes from the effects on long-term expansion of stem/progenitor cells. Gene expression profiling allowed the identification of GATA1-dependent and GATA1-independent STAT5 target genes, and these studies revealed that several proliferation-related genes were up-regulated by STAT5 independent of GATA1, whereas several erythroid differentiation-related genes were found to be GATA1 as well as STAT5 dependent.


Subject(s)
Erythropoiesis/genetics , Erythropoiesis/physiology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Down-Regulation , Fetal Blood/cytology , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/deficiency , Gene Expression Profiling , Humans , In Vitro Techniques , Infant, Newborn , Myelopoiesis/genetics , Myelopoiesis/physiology , RNA Interference , STAT5 Transcription Factor/genetics , Transduction, Genetic
14.
Proc Natl Acad Sci U S A ; 107(12): 5681-6, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20176930

ABSTRACT

To identify genetic factors that interact with social environments to impact human health, we used a bioinformatic strategy that couples expression array-based detection of environmentally responsive transcription factors with in silico discovery of regulatory polymorphisms to predict genetic loci that modulate transcriptional responses to stressful environments. Tests of one predicted interaction locus in the human IL6 promoter (SNP rs1800795) verified that it modulates transcriptional response to beta-adrenergic activation of the GATA1 transcription factor in vitro. In vivo validation studies confirmed links between adverse social conditions and increased transcription of GATA1 target genes in primary neural, immune, and cancer cells. Epidemiologic analyses verified the health significance of those molecular interactions by documenting increased 10-year mortality risk associated with late-life depressive symptoms that occurred solely for homozygous carriers of the GATA1-sensitive G allele of rs1800795. Gating of depression-related mortality risk by IL6 genotype pertained only to inflammation-related causes of death and was associated with increased chronic inflammation as indexed by plasma C-reactive protein. Computational modeling of molecular interactions, in vitro biochemical analyses, in vivo animal modeling, and human molecular epidemiologic analyses thus converge in identifying beta-adrenergic activation of GATA1 as a molecular pathway by which social adversity can alter human health risk selectively depending on individual genetic status at the IL6 locus.


Subject(s)
Interleukin-6/genetics , Social Environment , Base Sequence , Computational Biology , DNA/genetics , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , Humans , Models, Genetic , Molecular Epidemiology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Stress, Psychological , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Transcriptional Activation
15.
Int J Oncol ; 34(3): 853-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212691

ABSTRACT

The hematopoietic transcription factor GATA-1 regulates the expression of several genes associated with differentiation of erythroid cells. We show here the inhibitory effect of tumor necrosis factor alpha (TNFalpha), a proinflammatory cytokine, on hemoglobinization and erythroid transcription factor GATA-1 expression in erythroleukemia (HEL) as well as in chronic myelogenous leukemia (K562) cells, which were induced to differentiate towards the erythroid lineage after aclacinomycin (Acla), doxorubicin (Dox) or hemin (HM) treatment. As a result, we observed i) a decreased expression of Friend of GATA-1 (FOG-1), an essential cofactor of GATA-1 transcription factor, ii) a downregulation of GATA-1 by proteasomal degradation and iii) a reduced acetylation level of GATA-1 in HM-induced K562 cells after TNF treatment. As a result, these modifications i) decreased the level of GATA-1/FOG-1 complex, ii) unsettled the GATA-1/GATA-2 balance, iii) reduced GATA-1 transcriptional activity and iv) inhibited erythroid marker gene expression (glycophorin A, erythropoietin receptor, gamma-globin) independently of the cell line or the inducer used. These data provided new insights into the role of GATA-1 regulation in TNFalpha-mediated inhibition of erythroid differentiation in erythroleukemia.


Subject(s)
Erythroid Cells/drug effects , GATA1 Transcription Factor/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Aclarubicin/analogs & derivatives , Aclarubicin/pharmacology , Blotting, Western , Cell Differentiation/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Erythroid Cells/metabolism , Erythroid Cells/pathology , GATA1 Transcription Factor/biosynthesis , GATA1 Transcription Factor/genetics , Hemin/pharmacology , Hemoglobins/biosynthesis , Humans , K562 Cells , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
16.
Nucleic Acids Res ; 36(14): 4521-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18586828

ABSTRACT

GATA-1 is an erythroid activator that binds beta-globin gene promoters and DNase I hypersensitive sites (HSs) of the beta-globin locus control region (LCR). We investigated the direct role of GATA-1 interaction at the LCR HS2 enhancer by mutating its binding sites within minichromosomes in erythroid cells. Loss of GATA-1 in HS2 did not compromise interaction of NF-E2, a second activator that binds to HS2, nor was DNase I hypersensitivity at HS2 or the promoter of a linked epsilon-globin gene altered. Reduction of NF-E2 using RNAi confirmed the overall importance of this activator in establishing LCR HSs. However, recruitment of the histone acetyltransferase CBP and RNA pol II to HS2 was diminished by GATA-1 loss. Transcription of epsilon-globin was severely compromised with loss of RNA pol II from the transcription start site and reduction of H3 acetylation and H3K4 di- and tri-methylation in coding sequences. In contrast, widespread detection of H3K4 mono-methylation was unaffected by loss of GATA-1 in HS2. These results support the idea that GATA-1 interaction in HS2 has a prominent and direct role in co-activator and pol II recruitment conferring active histone tail modifications and transcription activation to a target gene but that it does not, by itself, play a major role in establishing DNase I hypersensitivity.


Subject(s)
GATA1 Transcription Factor/physiology , Globins/genetics , Locus Control Region , Trans-Activators/physiology , Transcriptional Activation , Acetylation , Binding Sites , CREB-Binding Protein/metabolism , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/metabolism , Histones/chemistry , Histones/metabolism , Humans , K562 Cells , Methylation , Mutation , NF-E2 Transcription Factor, p45 Subunit/antagonists & inhibitors , NF-E2 Transcription Factor, p45 Subunit/physiology , Open Reading Frames , RNA Polymerase II/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
17.
J Exp Med ; 205(3): 611-24, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18299398

ABSTRACT

The zinc finger transcription factor GATA-1 requires direct physical interaction with the cofactor friend of GATA-1 (FOG-1) for its essential role in erythroid and megakaryocytic development. We show that in the mast cell lineage, GATA-1 functions completely independent of FOG proteins. Moreover, we demonstrate that FOG-1 antagonizes the fate choice of multipotential progenitor cells for the mast cell lineage, and that its down-regulation is a prerequisite for mast cell development. Remarkably, ectopic expression of FOG-1 in committed mast cell progenitors redirects them into the erythroid, megakaryocytic, and granulocytic lineages. These lineage switches correlate with transcriptional down-regulation of GATA-2, an essential mast cell GATA factor, via switching of GATA-1 for GATA-2 at a key enhancer element upstream of the GATA-2 gene. These findings illustrate combinatorial control of cell fate identity by a transcription factor and its cofactor, and highlight the role of transcriptional networks in lineage determination. They also provide evidence for lineage instability during early stages of hematopoietic lineage commitment.


Subject(s)
GATA1 Transcription Factor/antagonists & inhibitors , GATA2 Transcription Factor/antagonists & inhibitors , Mast Cells/cytology , Mast Cells/metabolism , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , GATA1 Transcription Factor/deficiency , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/deficiency , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Yolk Sac/cytology , Yolk Sac/metabolism
18.
J Immunol ; 178(1): 360-8, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17182574

ABSTRACT

Mast cells are pivotal effector cells in IgE-mediated allergic reactions. GATA transcriptional factors such as GATA-1 and GATA-2 are expressed in mast cells, and recent studies have revealed that both GATA-1 and GATA-2 are required for mast cell development. However, the role of GATA transcriptional factors in differentiated mast cells has remained largely unknown. In this study, we repressed the activity of GATA-1 and GATA-2 by using three different approaches (inducible overexpression of a dominant-negative form of GATA, pharmacological inactivation, or small interfering RNA technology), and analyzed the molecular mechanisms of GATA transcriptional factors in the activation of mast cells. Surprisingly, the repression of GATA activity in differentiated mast cells led to the impairment of cell survival, IgE-induced degranulation, and cytokine production. Signal transduction and histone modification in the chromatin related to protein kinase Cbeta were defective in these cells. These results identify that GATA has a critical role in the activation of mast cell.


Subject(s)
GATA1 Transcription Factor/physiology , GATA2 Transcription Factor/physiology , Mast Cells/immunology , Acetylation , Animals , Cell Degranulation , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cytokines/metabolism , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/antagonists & inhibitors , GATA2 Transcription Factor/genetics , Gene Expression Profiling , Histones/metabolism , Immunoglobulin E/pharmacology , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Receptors, IgE/metabolism , Signal Transduction
19.
Blood ; 108(1): 262-9, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16522818

ABSTRACT

Cell-type-specific transcription of mouse high-affinity IgE receptor (FcepsilonRI) beta-chain is positively regulated by the transcription factor GATA-1. Although GATA-1 is expressed in erythroid cells, megakaryocytes, and mast cells, the expression of mouse FcepsilonRI beta-chain is restricted to mast cells. In the present study, we characterized the role of GATA-associated cofactor FOG-1 in the regulation of the FcepsilonRI beta-chain promoter. The expression levels of FOG-1, GATA-1, and beta-chain in each hematopoietic cell line were analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting. FOG-1 expression was higher in the beta-chain-negative hematopoietic progenitor cell line Ba/F3 than in the beta-chain-positive mast cell line PT18. By contrast, GATA-1 expression was similar when comparing the 2 cell lines. A transient reporter assay demonstrated that the beta-chain promoter functioned in PT18 but not in Ba/F3 and that the transcription activity of the beta-chain promoter in PT18 was markedly suppressed by overexpression of FOG-1. Although the activity of the beta-chain promoter, which was upregulated by coexpression of GATA-1, was significantly suppressed by coexpression of FOG-1 in the simian kidney CV-1 cells (beta-chain(-), GATA-1(-), and FOG-1(-)), the transactivation of the beta-chain promoter by the GATA-1 mutant V205G, which cannot bind FOG-1, was not affected by coexpression of FOG-1. Further, overexpression of FOG-1 in PT18 resulted in decreases in cell surface expression of FcepsilonRI and beta-chain transcription. Finally, suppression of FOG-1 expression using an siRNA approach resulted in increased beta-chain promoter activity in Ba/F3. These results suggest that FOG-1 expression level regulates the GATA-1-dependent FcepsilonRI beta-chain promoter.


Subject(s)
GATA1 Transcription Factor/antagonists & inhibitors , Gene Expression Regulation/genetics , Mast Cells/immunology , Nuclear Proteins/pharmacology , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/genetics , Transcription Factors/pharmacology , Transcription, Genetic , Animals , Cell Line , Down-Regulation/drug effects , Down-Regulation/immunology , GATA1 Transcription Factor/immunology , Gene Expression Regulation/drug effects , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/immunology , Receptors, IgE/immunology , Structure-Activity Relationship , Transcription Factors/biosynthesis , Transcription Factors/immunology
20.
Cancer Lett ; 240(2): 203-12, 2006 Aug 28.
Article in English | MEDLINE | ID: mdl-16274927

ABSTRACT

Up-regulation of tumor necrosis factor alpha (TNFalpha) is linked to solid tumors as well as to hematologic disorders including different forms of anemia and multiple myeloma. This cytokine was shown to contribute to inhibition of erythroid maturation mechanisms which are characterized by the expression of specific genes regulated by GATA-1 and NF-E2 transcription factors. Here, we assessed the inhibiting effect of TNFalpha on erythroid differentiation using K562 cells which can be chemically induced to differentiate towards the erythroid pathway by aclacinomycin A, an anthracyclin. Results show that induced hemoglobinization of K562 cells as well as gamma-globin and erythropoietin receptor gene expression are decreased by TNFalpha via the inhibition of GATA-1 at its mRNA and protein expression level. Additionally, both constitutive and induced binding activity of GATA-1 is abolished and induced activation of a GATA-1 driven luciferase reporter construct is inhibited. Altogether, our results provide insight into the molecular mechanisms of inflammation-induced inhibition of erythroid differentiation.


Subject(s)
Aclarubicin/pharmacology , Antibiotics, Antineoplastic/pharmacology , Cell Differentiation/drug effects , Erythroid Precursor Cells/drug effects , GATA1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Electrophoretic Mobility Shift Assay , GATA1 Transcription Factor/antagonists & inhibitors , GATA1 Transcription Factor/genetics , Globins/metabolism , Humans , K562 Cells/drug effects , Luciferases/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Erythropoietin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL