The GATA-1/FOG-1/NuRD Axis Is Essential for Normal Hematopoiesis.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 375-375
Author(s):  
Wei Hong ◽  
Hongxin Wang ◽  
Yuhuan Wang ◽  
John K. Choi ◽  
Suresh G. Shelat ◽  
...  

Abstract GATA-1 controls the development of erythroid cells and megakaryocytes through its ability to activate and repress gene transcription. GATA-1 binds many nuclear proteins, but only a few of these associations have been examined in vivo. One important example is FOG-1, a critical cofactor that contributes to both gene activation and repression by GATA-1. Loss of FOG-1 generally phenocopies GATA-1 deficiency, impairing both erythroid and megakaryocytic differentiation. We reported previously that FOG-1 directly binds the NuRD protein complex, which contains histone deacetylase and chromatin remodeling activities. This provides one mechanism for GATA-1/FOG-1-mediated gene repression. Accordingly, ChIP profiling of the NuRD proteins MTA-2, RbAp46 and Mi-2β revealed the presence of these molecules at the Kit and Gata2 genes both of which are directly repressed by GATA-1 in a FOG-1-dependent manner. NuRD proteins were spread broadly across the Kit and Gata2 genes but were further enriched at sites occupied by GATA-1 and FOG-1 in vivo. Unexpectedly, we also observed NuRD components at GATA-1-activated genes including β-globin and Ahsp. Moreover, the ability of FOG-1 to augment GATA-1-induced transcription in transient transfection assays required NuRD binding. Hence, NuRD may be bi-functional, contributing to either gene activation or repression, depending on the transcriptional and cellular context. To study the role of the FOG-1/NuRD interaction in vivo we generated mice bearing missense mutations in the Fog-1(Zfpm1) gene that disrupt NuRD binding in the FOG-1 protein. Homozygous mutant mice are born at reduced Mendelian ratios. Surviving animals display ineffective erythropoiesis marked by splenomegaly and impaired erythroid maturation. In addition, homozygous mutant animals display macrothrombocytopenia with impaired platelet function. Thus, recruitment of NuRD by GATA-1 and FOG-1 is essential for both erythropoiesis and megakaryocytopoiesis. Ongoing studies include further phenotypic analysis of the mutant mice, including comparative gene expression analysis in stage-matched wild-type and mutant erythroid cells to identify critical NuRD-dependent GATA target genes, and to resolve whether NuRD is essential for both activation and repression by GATA-1 and FOG-1 in vivo. An important open question under investigation is how recruitment of the NURD complex can lead to suppression of some genes and the enhanced expression of others. The FOG-1/NuRD mutant mice provide useful tools to dissect transcription pathways initiated by GATA-1. Moreover, given the role of GATA-1 mutations in congenital anemias and megakaryoblastic leukemias, enzymatic components of the NuRD complex may provide novel targets for pharmacologic manipulation to treat these disorders.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1459-1459
Author(s):  
Yi Luo ◽  
Jorg Bungert ◽  
Suming Huang ◽  
Yi Qiu

Abstract Abstract 1459 Poster Board I-482 Histone deacetylases (HDACs) play important roles in transcriptional regulation in eukaryotic cells. Several lines of evidence also link HDACs to cancer. HDAC inhibitors have shown preclinical promise when combined with other therapeutic agents in the treatments of leukemia and solid tumors. HDAC1 often presents in repressor complexes, such as Sin3, NuRD and CoREST complexes. We previously found that HDAC1 can be acetylated in vivo by acetyltransferase p300 and that acetylated HDAC1 completely loses its deacetylase activity (Qiu et al, 2006). In this study, we investigate the role of HDAC1 during erythroid differentiation. HDAC1 deacetylase activity gradually reduced upon the induction of differentiation. However, the HDAC1 protein level remains unchanged, suggesting that HDAC1 deacetylase activity, but not its protein level, is regulated. To further test the role of HDAC1 in erythroid differentiation, stable cell lines that overexpress HDAC1 and mutants mimicking acetylated or unacetylated HDAC1 were established. We also generated stable HDAC1 and HDAC2 knock down cell lines. The results showed that HDAC1 deacetylase activity is required to promote erythroid proliferation and to inhibit differentiation. Next, we studied whether HDAC1 modulates erythroid differentiation through regulating the activity of key erythroid transcription factor GATA-1. GATA-1 interacts with many other key hematopoietic transcription factors, as well as chromatin remodeling/modifying coactivators and corepressors. It is suggested that GATA-1 mediates gene activation through its association with coactivator complexes. However, recent studies indicated that GATA-1 associates with HDAC1/2 containing corepressor complexes (NuRD) throughout differentiation of erythroid cells. We investigated the deacetylase activity of the GATA-1 associated NuRD complex during erythroid differentiation. We found that the deacetylase activity of the complex decreased and further diminished at day 5 of DMSO induced MEL cells. Accordingly, the acetylated form of HDAC1 within the GATA-1 complex increased during erythroid differentiation. We further demonstrated the role of HDAC1 in GATA-1 mediated gene transcription in transient transfection assays. These studies indicate that HDAC1 plays an important role in regulating GATA-1 activity and the deacetylase activity of the GATA-1 associated NuRD complex is also regulated. This complex may play different roles in undifferentiated and differentiated erythroid cells. Thus, our results suggest a novel but rather general regulatory mechanism of histone deacetylase containing protein complexes. Disclosures No relevant conflicts of interest to declare.


2004 ◽  
Vol 24 (20) ◽  
pp. 9026-9037 ◽  
Author(s):  
Daniel R. Buchholz ◽  
Akihiro Tomita ◽  
Liezhen Fu ◽  
Bindu D. Paul ◽  
Yun-Bo Shi

ABSTRACT Thyroid hormone (T3) has long been known to be important for vertebrate development and adult organ function. Whereas thyroid hormone receptor (TR) knockout and transgenic studies of mice have implicated TR involvement in mammalian development, the underlying molecular bases for the resulting phenotypes remain to be determined in vivo, especially considering that T3 is known to have both genomic, i.e., through TRs, and nongenomic effects on cells. Amphibian metamorphosis is an excellent model for studying the role of TR in vertebrate development because of its total dependence on T3. Here we investigated the role of TR in metamorphosis by developing a dominant positive mutant thyroid hormone receptor (dpTR). In the frog oocyte transcription system, dpTR bound a T3-responsive promoter and activated the promoter independently of T3. Transgenic expression of dpTR under the control of a heat shock-inducible promoter in premetamorphic tadpoles led to precocious metamorphic transformations. Molecular analyses showed that dpTR induced metamorphosis by specifically binding to known T3 target genes, leading to increased local histone acetylation and gene activation, similar to T3-bound TR during natural metamorphosis. Our experiments indicated that the metamorphic role of T3 is through genomic action of the hormone, at least on the developmental parameters tested. They further provide the first example where TR is shown to mediate directly and sufficiently these developmental effects of T3 in individual organs by regulating target gene expression in these organs.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3443-3443
Author(s):  
AI Inoue ◽  
Tohru Fujiwara ◽  
Yoko Okitsu ◽  
Noriko Fukuhara ◽  
Yasushi Onishi ◽  
...  

Abstract Abstract 3443 Background: Developmental control mechanisms often utilize multimeric complexes containing transcription factors, coregulators, and additional non-DNA binding components. It is challenging to ascertain how such components contribute to complex function at endogenous loci. LMO2 (LIM-only protein 2) is a non-DNA binding transcriptional coregulator, and is an important regulator of hematopoietic stem cell development and erythropoiesis, as mice lacking this gene show defects in blood formation as well as fetal erythropoiesis (Warren et al. Cell. 1994). In the context of erythropoiesis, LMO2 has been demonstrated to be a part of multimetric complex, including master regulators of hematopoiesis (GATA-1 and SCL/TAL1), chromatin looping factor LDB1 and hematopoietic corepressor ETO2 (referred as GATA-SCL/TAL1 complex). As LMO2 controls hematopoiesis, its dysregulation is leukemogenic, and its influence on GATA factor function is still not evident, we investigated here the transcriptional regulatory mechanism via LMO2 in erythroid cells. Methods: For LMO2 knockdown, anti-LMO2 siRNA (Thermo Scientific Dharmacon) and pGIPZ lentiviral shRNAmir system (Open Biosystems) were used. Western blotting and Quantitative ChIP analysis were performed using antibodies for GATA-1, LMO2 (abcam), GATA-2, TAL1 and LDB1 (Santa Cruz). To obtain human primary erythroblasts, CD34-positive cells isolated from cord blood were induced in liquid suspension culture. For transcription profiling, human whole expression array was used (Agilent), and the data was analyzed with GeneSpring GX software. To induce erythroid differentiation of K562 cells, hemin was treated at a concentration of 30 uM for 24h. Results: siRNA-mediated LMO2 knockdown in hemin-treated K562 cells results in significantly decreased ratio of benzidine-staining positive cells, suggesting that LMO2 has an important role in the erythroid differentiation of K562 cells. Next, we conducted microarray analysis to characterize LMO2 target gene ensemble in K562 cells. In contrast to the predominantly repressive role of LMO2 in murine G1E-ER-GATA-1 cells (Fujiwara et al. PNAS. 2010), the analyses (n = 2) demonstrated that 177 and 78 genes were upregulated and downregulated (>1.5-fold), respectively, in the LMO2-knockdowned K562 cells. Downregulated gene ensemble contained prototypical erythroid genes such as HBB and SLC4A1 (encodes erythrocyte membrane protein band 3). To test what percentages of LMO2-regulated genes could be direct target genes of GATA-1 in K562 cells, we merged the microarray results with ChIP-seq profile (n= 5,749, Fujiwara et al. Mol Cell. 2009), and demonstrated that 26.4% and 23.1% of upregulated and downregulated genes, respectively, contained significant GATA-1 peaks in their loci. Furthermore, whereas LMO2 knockdown in K562 cells did not affect the expression of GATA-1, GATA-2 and SCL/TAL1 based on quantitative RT-PCR as well as Western blotting, the knockdown resulted in the significantly decreased chromatin occupancy of GATA-1, GATA-2, SCL/TAL1 and LDB1 at beta-globin locus control region and SLC4A1 locus. We subsequently analyzed the consequences of LMO2 knockdown in primary erythroblasts. Endogeneous LMO2 expression was upregulated along with the differentiation of cord blood cell-derived primary erythroblasts. shRNA-mediated knockdown of LMO2 in primary erythroblasts resulted in significant downregulation of HBB, HBA and SLC4A1. Conclusion: Our results suggest that LMO2 contributes to the expression of GATA-1 target genes in a context-dependent manner, through modulating the assembly of the components of GATA-SCL/TAL1 complex at endogeneous loci. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 24 (17) ◽  
pp. 7548-7558 ◽  
Author(s):  
Akihira Ohtoshi ◽  
Richard R. Behringer

ABSTRACT Dmbx1 encodes a paired-like homeodomain protein that is expressed in developing neural tissues during mouse embryogenesis. To elucidate the in vivo role of Dmbx1, we generated two Dmbx1 mutant alleles. Dmbx1− lacks the homeobox and Dmbx1z is an insertion of a lacZ reporter gene. Dmbx1z appears to be a faithful reporter of Dmbx1 expression during embryogenesis and after birth. Dmbx1-lacZ expression was detected in the superior colliculus, cerebellar nuclei, and subpopulations of the medulla oblongata and spinal cord. Some Dmbx1 homozygous mutant mice died during the neonatal period, while others survived to adulthood; however, their growth was impaired. Both heterozygous and homozygous mutant offspring from Dmbx1 homozygous mutant females exhibited a low survival rate and poor growth. However, even wild-type pups fostered onto Dmbx1 homozygous mutant females grew poorly, suggesting a Dmbx1-dependent nursing defect. Dmbx1 mutant mice had an aberrant Dmbx1-lacZ expression pattern in the nervous system, indicating that they had abnormal brain development. These results demonstrate that Dmbx1 is required for postnatal survival, growth, and brain development.


2019 ◽  
Vol 216 (6) ◽  
pp. 1431-1449 ◽  
Author(s):  
Hiroki Tanaka ◽  
Yasunobu Arima ◽  
Daisuke Kamimura ◽  
Yuki Tanaka ◽  
Noriyuki Takahashi ◽  
...  

Regnase-1 (also known as Zc3h12a or MCPIP-1) is an endoribonuclease involved in mRNA degradation of inflammation-associated genes. Regnase-1 is inactivated in response to external stimuli through post-translational modifications including phosphorylation, yet the precise role of phosphorylation remains unknown. Here, we demonstrate that interleukin (IL)-17 induces phosphorylation of Regnase-1 in an Act1-TBK1/IKKi–dependent manner, especially in nonhematopoietic cells. Phosphorylated Regnase-1 is released from the endoplasmic reticulum (ER) into the cytosol, thereby losing its mRNA degradation function, which leads to expression of IL-17 target genes. By using CRISPR/Cas-9 technology, we generated Regnase-1 mutant mice, in which IL-17–induced Regnase-1 phosphorylation is completely blocked. Mutant mice (Regnase-1AA/AA and Regnase-1ΔCTD/ΔCTD) were resistant to the IL-17–mediated inflammation caused by T helper 17 (Th17) cells in vivo. Thus, Regnase-1 plays a critical role in the development of IL-17–mediated inflammatory diseases via the Act1-TBK1-IKKi axis, and blockade of Regnase-1 phosphorylation sites may be promising for treatment of Th17-associated diseases.


Blood ◽  
2008 ◽  
Vol 112 (9) ◽  
pp. 3889-3899 ◽  
Author(s):  
David Garrick ◽  
Marco De Gobbi ◽  
Vasiliki Samara ◽  
Michelle Rugless ◽  
Michelle Holland ◽  
...  

Although much is known about globin gene activation in erythroid cells, relatively little is known about how these genes are silenced in nonerythroid tissues. Here we show that the human α- and β-globin genes are silenced by fundamentally different mechanisms. The α-genes, which are surrounded by widely expressed genes in a gene dense region of the genome, are silenced very early in development via recruitment of the Polycomb (PcG) complex. By contrast, the β-globin genes, which lie in a relatively gene-poor chromosomal region, are not bound by this complex in nonerythroid cells. The PcG complex seems to be recruited to the α-cluster by sequences within the CpG islands associated with their promoters; the β-globin promoters do not lie within such islands. Chromatin associated with the α-globin cluster is modified by histone methylation (H3K27me3), and silencing in vivo is mediated by the localized activity of histone deacetylases (HDACs). The repressive (PcG/HDAC) machinery is removed as hematopoietic progenitors differentiate to form erythroid cells. The α- and β-globin genes thus illustrate important, contrasting mechanisms by which cell-specific hematopoietic genes (and tissue-specific genes in general) may be silenced.


2017 ◽  
Vol 8 (1) ◽  
Author(s):  
Gongwei Wu ◽  
Mengqiu Yuan ◽  
Shengqi Shen ◽  
Xiaoyu Ma ◽  
Jingwen Fang ◽  
...  

Abstract Menin is an enigmatic protein that displays unique ability to either suppress or promote tumorigenesis in a context-dependent manner. The role for Menin to promote oncogenic functions has been largely attributed to its essential role in forming the MLL methyltransferase complex, which mediates H3K4me3. Here, we identify an unexpected role of Menin in enhancing the transactivity of oncogene MYC in a way independent of H3K4me3 activity. Intriguingly, we find that Menin interacts directly with the TAD domain of MYC and co-localizes with MYC to E-Box to enhance the transcription of MYC target genes in a P-TEFb-dependent manner. We further demonstrate that, by transcriptionally promoting the expression of MYC target genes in cancer cells, Menin stimulates cell proliferation and cellular metabolism both in vitro and in vivo. Our results uncover a previously unappreciated mechanism by which Menin functions as an oncogenic regulatory factor that is critical for MYC-mediated gene transcription.


2016 ◽  
Vol 75 (11) ◽  
pp. 2037-2044 ◽  
Author(s):  
Neng-Yu Lin ◽  
Alfiya Distler ◽  
Christian Beyer ◽  
Ariella Philipi-Schöbinger ◽  
Silvia Breda ◽  
...  

ObjectivesNotch ligands and receptors have recently been shown to be differentially expressed in osteoarthritis (OA). We aim to further elucidate the functional role of Notch signalling in OA using Notch1 antisense transgenic (Notch1 AS) mice.MethodsNotch and hedgehog signalling were analysed by real-time PCR and immunohistochemistry. Notch-1 AS mice were employed as a model of impaired Notch signalling in vivo. Experimental OA was induced by destabilisation of the medial meniscus (DMM). The extent of cartilage destruction and osteophyte formation was analysed by safranin-O staining with subsequent assessment of the Osteoarthritis Research Society International (OARSI) and Mankin scores and µCT scanning. Collagen X staining was used as a marker of chondrocyte hypertrophy. The role of hairy/enhancer of split 1 (Hes-1) was investigated with knockdown and overexpression experiments.ResultsNotch signalling was activated in human and murine OA with increased expression of Jagged1, Notch-1, accumulation of the Notch intracellular domain 1 and increased transcription of Hes-1. Notch1 AS mice showed exacerbated OA with increases in OARSI scores, osteophyte formation, increased subchondral bone plate density, collagen X and osteocalcin expression and elevated levels of Epas1 and ADAM-TS5 mRNA. Inhibition of the Notch pathway induced activation of hedgehog signalling with induction of Gli-1 and Gli-2 and increased transcription of hedgehog target genes. The regulatory effects of Notch signalling on Gli-expression were mimicked by Hes-1.ConclusionsInhibition of Notch signalling activates hedgehog signalling, enhances chondrocyte hypertrophy and exacerbates experimental OA including osteophyte formation. These data suggest that the activation of the Notch pathway may limit aberrant hedgehog signalling in OA.


2008 ◽  
Vol 29 (5) ◽  
pp. 1321-1337 ◽  
Author(s):  
Prabhat Kumar Purbey ◽  
Sunita Singh ◽  
Dimple Notani ◽  
P. Pavan Kumar ◽  
Amita S. Limaye ◽  
...  

ABSTRACT Special AT-rich binding protein 1 (SATB1) acts as a global regulator of gene expression by recruiting various corepressor or coactivator complexes, thereby establishing a unique chromatin structure at its genomic targets in a context-dependent manner. Although SATB1 acts predominantly as a repressor via recruitment of histone deacetylase 1 (HDAC1) complexes, the precise mechanism of global repression is not clear. Here we report that SATB1 and C-terminal binding protein 1 (CtBP1) form a repressor complex in vivo. The interaction occurs via the CtBP1 interaction consensus motif PVPLS within the PDZ-like domain of SATB1. The acetylation of SATB1 upon LiCl and ionomycin treatments disrupts its association with CtBP1, resulting in enhanced target gene expression. Chromatin immunoprecipitation analysis indicated that the occupancy of CtBP1 and HDAC1 is gradually decreased and the occupancy of PCAF is elevated at the SATB1 binding sites within the human interleukin-2 and mouse c-Myc promoters. Moreover, gene expression profiling studies using cells in which expression of SATB1 and CtBP1 was silenced indicated commonly targeted genes that may be coordinately repressed by the SATB1-CtBP1 complex. Collectively, these results provide a mechanistic insight into the role of SATB1-CtBP1 interaction in the repression and derepression of SATB1 target genes during Wnt signaling in T cells.


1999 ◽  
Vol 380 (7-8) ◽  
pp. 953-959 ◽  
Author(s):  
P. O. Hassa ◽  
M. O. Hottiger

AbstractThe transcription factor NF-κB plays a critical role in immune and inflammatory responses. Here we show that poly (ADP ribose) polymerase (PARP) is required for specific NF-κB transcriptional activationin vivo. The activation of the HIV-LTR promoter and an NF-κBdependent artificial promoter was drastically reduced in PARP (_/_) cells, independently of the signaling pathway through which NF-bB was induced. Furthermore NF-κB-dependent gene activation was restoredin vivoby the expression of PARP in PARP (_/_) cells. Finally, we show that both NF-κB and PARP formed a stable immunoprecipitable nuclear complex. This interaction did not need DNA binding. Our results suggest that PARP is an important cofactor in the activation cascade of NF-κB-dependent target genes.


Sign in / Sign up

Export Citation Format

Share Document