scholarly journals Lysine acetyltransferase Tip60 is required for hematopoietic stem cell maintenance

Blood ◽  
2020 ◽  
Vol 136 (15) ◽  
pp. 1735-1747 ◽  
Author(s):  
Akihiko Numata ◽  
Hui Si Kwok ◽  
Qi-Ling Zhou ◽  
Jia Li ◽  
Roberto Tirado-Magallanes ◽  
...  

Abstract Hematopoietic stem cells (HSCs) have the potential to replenish the blood system for the lifetime of the organism. Their 2 defining properties, self-renewal and differentiation, are tightly regulated by the epigenetic machineries. Using conditional gene-knockout models, we demonstrated a critical requirement of lysine acetyltransferase 5 (Kat5, also known as Tip60) for murine HSC maintenance in both the embryonic and adult stages, which depends on its acetyltransferase activity. Genome-wide chromatin and transcriptome profiling in murine hematopoietic stem and progenitor cells revealed that Tip60 colocalizes with c-Myc and that Tip60 deletion suppress the expression of Myc target genes, which are associated with critical biological processes for HSC maintenance, cell cycling, and DNA repair. Notably, acetylated H2A.Z (acH2A.Z) was enriched at the Tip60-bound active chromatin, and Tip60 deletion induced a robust reduction in the acH2A.Z/H2A.Z ratio. These results uncover a critical epigenetic regulatory layer for HSC maintenance, at least in part through Tip60-dependent H2A.Z acetylation to activate Myc target genes.

Blood ◽  
2012 ◽  
Vol 120 (10) ◽  
pp. 2076-2086 ◽  
Author(s):  
Britta Will ◽  
Li Zhou ◽  
Thomas O. Vogler ◽  
Susanna Ben-Neriah ◽  
Carolina Schinke ◽  
...  

Abstract Even though hematopoietic stem cell (HSC) dysfunction is presumed in myelodysplastic syndrome (MDS), the exact nature of quantitative and qualitative alterations is unknown. We conducted a study of phenotypic and molecular alterations in highly fractionated stem and progenitor populations in a variety of MDS subtypes. We observed an expansion of the phenotypically primitive long-term HSCs (lineage−/CD34+/CD38−/CD90+) in MDS, which was most pronounced in higher-risk cases. These MDS HSCs demonstrated dysplastic clonogenic activity. Examination of progenitors revealed that lower-risk MDS is characterized by expansion of phenotypic common myeloid progenitors, whereas higher-risk cases revealed expansion of granulocyte-monocyte progenitors. Genome-wide analysis of sorted MDS HSCs revealed widespread methylomic and transcriptomic alterations. STAT3 was an aberrantly hypomethylated and overexpressed target that was validated in an independent cohort and found to be functionally relevant in MDS HSCs. FISH analysis demonstrated that a very high percentage of MDS HSC (92% ± 4%) carry cytogenetic abnormalities. Longitudinal analysis in a patient treated with 5-azacytidine revealed that karyotypically abnormal HSCs persist even during complete morphologic remission and that expansion of clonotypic HSCs precedes clinical relapse. This study demonstrates that stem and progenitor cells in MDS are characterized by stage-specific expansions and contain epigenetic and genetic alterations.


2020 ◽  
Vol 21 (22) ◽  
pp. 8448
Author(s):  
Chun-Hao Hung ◽  
Keh-Yang Wang ◽  
Yae-Huei Liou ◽  
Jing-Ping Wang ◽  
Anna Yu-Szu Huang ◽  
...  

Erythroid Krüppel-like factor (EKLF/KLF1) was identified initially as a critical erythroid-specific transcription factor and was later found to be also expressed in other types of hematopoietic cells, including megakaryocytes and several progenitors. In this study, we have examined the regulatory effects of EKLF on hematopoiesis by comparative analysis of E14.5 fetal livers from wild-type and Eklf gene knockout (KO) mouse embryos. Depletion of EKLF expression greatly changes the populations of different types of hematopoietic cells, including, unexpectedly, the long-term hematopoietic stem cells Flk2− CD34− Lin− Sca1+ c-Kit+ (LSK)-HSC. In an interesting correlation, Eklf is expressed at a relatively high level in multipotent progenitor (MPP). Furthermore, EKLF appears to repress the expression of the colony-stimulating factor 2 receptor β subunit (CSF2RB). As a result, Flk2− CD34− LSK-HSC gains increased differentiation capability upon depletion of EKLF, as demonstrated by the methylcellulose colony formation assay and by serial transplantation experiments in vivo. Together, these data demonstrate the regulation of hematopoiesis in vertebrates by EKLF through its negative regulatory effects on the differentiation of the hematopoietic stem and progenitor cells, including Flk2− CD34− LSK-HSCs.


Author(s):  
Chong Zhang ◽  
Rui Huang ◽  
Xirui Ma ◽  
Jiehui Chen ◽  
Xinlu Han ◽  
...  

Ribosome biogenesis is a fundamental activity in cells. Ribosomal dysfunction underlies a category of diseases called ribosomopathies in humans. The symptomatic characteristics of ribosomopathies often include abnormalities in craniofacial skeletons, digestive organs, and hematopoiesis. Consistently, disruptions of ribosome biogenesis in animals are deleterious to embryonic development with hypoplasia of digestive organs and/or impaired hematopoiesis. In this study, ltv1, a gene involved in the small ribosomal subunit assembly, was knocked out in zebrafish by clustered regularly interspaced short palindromic repeats (CRISPRs)/CRISPR associated protein 9 (Cas9) technology. The recessive lethal mutation resulted in disrupted ribosome biogenesis, and ltv1Δ14/Δ14 embryos displayed hypoplastic craniofacial cartilage, digestive organs, and hematopoiesis. In addition, we showed that the impaired cell proliferation, instead of apoptosis, led to the defects in exocrine pancreas and hematopoietic stem and progenitor cells (HSPCs) in ltv1Δ14/Δ14 embryos. It was reported that loss of function of genes associated with ribosome biogenesis often caused phenotypes in a P53-dependent manner. In ltv1Δ14/Δ14 embryos, both P53 protein level and the expression of p53 target genes, Δ113p53 and p21, were upregulated. However, knockdown of p53 failed to rescue the phenotypes in ltv1Δ14/Δ14 larvae. Taken together, our data demonstrate that LTV1 ribosome biogenesis factor (Ltv1) plays an essential role in digestive organs and hematopoiesis development in zebrafish in a P53-independent manner.


2021 ◽  
Author(s):  
Elizabeth D. Howell ◽  
Amanda D. Yzaguirre ◽  
Peng Gao ◽  
Raphael Lis ◽  
Bing He ◽  
...  

Hematopoietic stem and progenitor cells (HSPCs) are generated de novo in the embryo from hemogenic endothelial cells (HECs) via an endothelial-to-hematopoietic transition (EHT) that requires the transcription factor RUNX1. Ectopic expression of RUNX1 alone can efficiently promote EHT and HSPC formation from embryonic endothelial cells (ECs), but less efficiently from fetal or adult ECs. Efficiency correlated with baseline accessibility of TGFβ-related genes associated with endothelial-to-mesenchymal transition (EndoMT) and participation of AP-1 and SMAD2/3 to initiate further chromatin remodeling along with RUNX1 at these sites. Activation of TGFβ signaling improved the efficiency with which RUNX1 specified fetal ECs as HECs. Thus, the ability of RUNX1 to promote EHT depends on its ability to recruit the TGFβ signaling effectors AP-1 and SMAD2/3, which in turn is determined by the changing chromatin landscape in embryonic versus fetal ECs. This work provides insight into regulation of EndoMT and EHT that will guide reprogramming efforts for clinical applications.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 371-371
Author(s):  
Ryohichi Sugimura ◽  
Areum Han ◽  
Deepak Jha ◽  
Yi-Fen Lu ◽  
Jeremy A Goettel ◽  
...  

Abstract A variety of tissues can be differentiated from pluripotent stem cells (PSCs) in vitro through stepwise exposure to morphogens, or by conversion of one differentiated cell type into another by enforced expression of master transcription factors (TFs). Despite considerable effort, neither approach has yielded functional human hematopoietic stem cells (HSCs). Building upon recent evidence that HSCs derive from definitive hemogenic endothelium (HE), we performed morphogen-directed differentiation of human PSCs into HE followed by screening of 26 candidate HSC-specifying TFs for the capacity to promote multi-lineage hematopoietic engraftment in irradiated immune deficient murine hosts. From genomic PCR of engrafted cells, we recovered seven TFs (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1, SPI1) that were sufficient to convert HE into hematopoietic stem and progenitor cells (HSPCs) that engraft GLY-A+ erythrocytes, CD33+ myeloid, CD15+ CD31+ neutrophils, CD19+ IgM+ B and CD3+ T cells in primary and secondary murine recipients for 12-14 weeks. Limiting dilution analysis indicated that the frequency of repopulating cells generated by this method was 1 in 4,707-15,029, lower than the frequency in CD34+ cord blood cells (1 in 1,819-5,173). Functional characterization of terminally differentiated cells demonstrated features of definitive erythropoiesis (expression of adult beta globin and enucleation). Engrafted neutrophils responded to cytokine stimuli by activation of myeloperoxidase. Human IgM and IgG could be detected in the serum of engrafted mice, and titers of ovalbumin specific antibody increased in response to protein immunization, indicating boostable immunity. T-cells responded to PMA/Ionomycin stimuli by activation of IFNγ, and sequencing of the T cell receptor revealed a broad clonotype diversity. Proviral integration analysis demonstrated derivation of myeloid and lymphoid progeny from common clones in secondary animals, indicating generation of self-renewing, multipotential HSC-like cells from PSCs. Mechanistically, the seven TFs induced HOXA target genes (LMO2, SOX4, MEIS1 and ID2); upregulated expression of homing-related genes (CXCR4, VLA5 and S1PR1); and enhanced the endothelial to hematopoietic transition (EHT), as indicated by a 2.4-fold induction of a RUNX1c-reporter. Our combined approach of morphogen-driven differentiation and TF-mediated cell fate conversion produced HSPCs from PSCs that hold promise for modeling hematopoietic disease in humanized mice and for therapeutic strategies in genetic blood disorders. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 714-714
Author(s):  
Jennifer Grants ◽  
Joanna Wegrzyn ◽  
David Knapp ◽  
Tony Hui ◽  
Kieran O'Neill ◽  
...  

Abstract MicroRNA miR-146a is frequently depleted in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Loss of miR-146a may be an initiating event in tumorigenesis, as miR-146a loss in mouse models is sufficient to cause features of MDS and eventual progression to AML. To define how miR-146a loss initiates tumorigenesis, we analyzed hematopoietic stem cell (HSC) function from miR-146a knockout (KO) mice prior to onset of an overt malignant phenotype. Tracking cell division kinetics, proliferation, and differentiation of single long-term HSC (LT-HSC; EPCR+CD45+CD48-CD150+) in culture, we found evidence that miR-146a KOreduces HSC quiescence and promotes differentiating cell divisions. Our data show that miR-146a KO HSC dysfunction may stem from loss of a CD150-bright EPCR-bright sub-population, which has previously been associated with robust HSC activity. In line with this, single cell DNA methylation profiling revealed a reduction in a primitive sub-population of LT-HSCs in miR-146a KO animals. In addition, single cell LT-HSC transplants revealed a myeloid repopulation bias. As reduced HSC cell cycle quiescence has been linked to impaired HSC self-renewal upon hematopoietic stress, such as serial transplantation, we assessed the frequency of serially transplantable HSCs by performing secondary transplants with limiting dilution. Serially transplantable HSC frequency was reduced in miR-146a KO compared to wild type, suggesting impaired HSC self-renewal. Transcriptome profiling of miR-146a KO hematopoietic stem and progenitor cells identified tumor necrosis factor (TNF) signaling activation as a potential driver of HSC dysfunction. LT-HSC cell cycle quiescence and the CD150-bright EPCR-bright LT-HSC sub-population were restored in miR-146a/TNF double KO mice, suggesting that aberrant TNF signaling activation drives HSC dysfunction upon loss of miR-146a. Gene expression levels in the TNF signaling network are inversely correlated with miR-146a levels in human AML, implying that TNF signaling may similarly disrupt HSC function in miR-146a- depleted myeloid malignancies. Overall, our findings suggest that miR-146a promotes HSC cell cycle quiescence and inhibits differentiation by antagonizing TNF signaling, in order to maintain a primitive sub-population of long-term self-renewing HSCs. Disclosures Eaves: Experimental Hematology: Other: Editor of journal; StemCell Technologies Inc: Other: Wife of owner.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 100-100
Author(s):  
Daisuke Shinoda ◽  
Yaeko Nakajima-Takagi ◽  
Motohiko Oshima ◽  
Atsunori Saraya ◽  
Hironori Harada ◽  
...  

Abstract Introduction: PcG proteins form two main multiprotein complexes, Polycomb repressive complex 1 (PRC1) and PRC2. They repress the transcription of target genes. Polycomb group ring finger protein1 (PCGF1) is a component of PRC1.1, a non-canonical PRC1.1 that monoubiquitylates H2A at lysine 119 in a manner independent of H3K27me3. Several groups including ours showed that the loss of Ezh2, a component of PRC2, promotes the development of JAK2 V617F-induced Myelofibrosis (MF) in mice. However, the role of PRC1.1 in hematologic malignancies is still not fully understood. We found that the deletion of PCGF1 in mice promotes myeloid commitment of hematopoietic stem and progenitor cells (HSPCs), and eventually induces a lethal myeloproliferative neoplasm (MPN)-like disease in mice (Nakajima-Takagi Y, unpublished data). Based on these findings, we investigated the role of PCGF1 in a mouse model of JAK2V617F-induced myelofibrosis. Methods: We transplanted BM cells from Cre-ERT2, PCGF1flox/flox;Cre-ERT2, JAK2V617F;Cre-ERT2, and JAK2V617F;PCGF1flox/flox;Cre-ERT2 mice into lethally irradiated recipient mice. We deleted PCGF1 by tamoxifen administration 4 weeks after transplantation. Results: JAK2/PCGF1 KO mice developed lethal MF significantly earlier than the other genotypes (p<0.01). JAK2/PCGF1 KO mice showed progressive anemia and severe thrombocytopenia. Bone marrow analysis of JAK2/PCGF1 KO mice revealed a significant reduction in HSPCs and an increase in the number of granulocyte-macrophage progenitors (GMPs). Erythropoiesis was severely impaired at the later stages of erythroid differentiation. To understand the molecular basis of MF-initiating cells in JAK2/PCGF1 KO mouse, we performed a gene expression analysis of LSKs/GMPs/MEPs isolated from the primary recipients 1 month after TAM injection. Gene set enrichment analysis of RNA-seq data clearly showed de-repression of PRC1 target genes marked with H2AK119ub1 in hematopoietic stem and progenitor cells (HSPCs) from JAK2/PCGF1 KO mice. The gene set of megakaryocyte progenitors was also positively enriched in JAK2/PCGF1 KO HSPCs. ChIP sequencing of H2AK119Ub revealed that the levels of H2AK119Ub at promoter regions were mildly reduced in JAK2/PCGF1 KO LK cells compared with Pcgf1 KO LK cells. Among differentially expressed genes, we found that HoxA cluster genes were de-repressed in JAK2/PCGF1 KO progenitor cells including MEPs following significant reductions in H2AK119Ub levels at the promoter regions. Lin28b-Let-7-Hmga2 pathway genes that are activated in JAK2/Ezh2 KO progenitor cells were not altered in expression in JAK2/PCGF1 KO progenitor cells, suggesting different mechanisms operating in the pathogenesis of JAK2/Ezh2 KO and JAK2/PCGF1 KO MF. A selective AURKA inhibitor has been reported to promote differentiation of megakaryocytes with PMF-associated mutations and had potent antifibrotic and antitumor activity in vivo in mouse models of PMF (Wen et al., Nat Med 21:1473, 2015). Following this report, we treated JAK2/PCGF1 KO mice with JAK inhibitors and/or AURKA inhibitors. Both inhibitors improved MF-related phenotypes including impaired erythroid differentiation of JAK2/PCGF1 KO mice. Conclusions: Our findings suggest that dysregulated PRC1.1 function promotes JAK2V617F-induced MF with mechanisms distinct from MF associated with PRC2 dysfunction. Disclosures Harada: Celgene: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3673-3673
Author(s):  
Kentson Lam ◽  
Randal Du ◽  
Shinobu Matsuura ◽  
Dong-Er Zhang

Abstract RUNX1, also known as AML1, is a DNA binding transcription factor that is expressed in hematopoietic stem and progenitor cells (HSPCs). As demonstrated by several mouse models, RUNX1 is necessary for definitive hematopoiesis and proper homeostasis of HSPCs. Furthermore, mutations of RUNX1have been implicated in patients with a variety of blood-related malignancies and disorders. These findings have established RUNX1 as a master regulator of hematopoiesis. As a transcription factor, RUNX1 exerts its function in hematopoiesis by binding to regulatory regions in order to guide the expression of its direct target genes. Most confirmed RUNX1 target genes are mainly expressed in differentiated blood cells. Direct targets of RUNX1 in HSPCs, however, have largely remained unexplored. Identifying direct target genes of RUNX1 offers an insightful view of how this master regulator influences HSPC function. To elucidate RUNX1 target genes in HSPCs, we have analyzed gene expression signatures from wildtype and RUNX1-deficient HSPCs (Lineage-/cKit+/Sca1+) in a previous report (Matsuura et al., Blood, 2012). With the goal of continuing the characterization of RUNX1 target genes, in this current study, we performed genome-occupancy analysis with chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) using RUNX1 antibodies and a murine HSPC cell line. Bioinformatics analysis of the ChIP-seq data revealed 6370 significant RUNX1 binding peaks (<1% FDR). The majority of these peaks were located in areas outside of promoter regions. The top de novo generated sequence motif from these peaks corresponds with the known RUNX binding consensus motif TG(T/C)GGT, suggesting that our ChIP-seq dataset is highly reliable. The combination of differential gene expression and RUNX1 genome occupancy data has revealed a list of candidate RUNX1-regulated target genes. We hypothesize that RUNX1 directly modulates the expression of these genes in normal hematopoiesis. One of the genes identified is Hmga2. We observed three RUNX1 binding peaks in the upstream, intron, and downstream regions relative to the Hmga2 gene locus. Furthermore, we confirmed strong up-regulation of Hmga2 in RUNX1-deficient HSPCs using reverse transcription coupled with quantitative polymerase chain reaction. HMGA2, also known as High Mobility Group AT-hook 2, is a non-histone chromatin protein. Its expression is highest during embryonic development and in undifferentiated cells. Over-expression of HMGA2 in transgenic mice or in bone marrow transplantation models have been reported to cause expansion of HSPCs. These reports indicate that HMGA2 is a significant mediator of HSPC proliferation. Interestingly, a major characteristic of mice without RUNX1 in their hematopoietic cells is the expansion of HSPCs, suggesting that HMGA2 may contribute to this phenotype. To further validate Hmga2 as a RUNX1 target gene, we cloned the Hmga2 promoter sequence and DNA fragments corresponding to the three RUNX1 binding peaks into luciferase reporter constructs and performed transfection studies using K562 and 293T cells. Interestingly, while these studies demonstrated strong responses to RUNX1 in promoter-luciferase assays, the effect of RUNX1 on Hmga2 promoter activity in these two cell lines was opposite. In addition, eliminating two RUNX binding motifs in the Hmga2 promoter did not affect RUNX1-mediated promoter-luciferase activity, indicating that there are additional regulatory mechanisms that may be important for RUNX1’s effect on the Hmga2 promoter. To examine the function of the three regions containing RUNX1 binding peaks in the Hmga2 gene locus, we also used luciferase reporter constructs including these regions in transfection studies. Increase of transcriptional activity was detected in the presence of the two regions that were upstream and downstream of the Hmga2 gene, suggesting that RUNX1 can act as a positive regulator through these regions. In contrast, the RUNX1 binding fragment in the intron region of Hmga2 reduced promoter-luciferase activity. This outcome indicates that RUNX1 acts as a suppressor through the Hmga2 intron element. In summary, these results establish Hmga2 as a novel RUNX1 target gene in HSPCs and mark the first study of the transcriptional regulation of the Hmga2 gene by RUNX1 through differential control regions. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document