Iron Restriction Via PKC Signaling Critically Contributes to Erythroid PU.1 Dysregulation in Anemia of Chronic Inflammation

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3451-3451
Author(s):  
Chante Richardson ◽  
Lorrie L. Delehanty ◽  
Adam Goldfarb

Abstract Abstract 3451 In addressing factors that suppress erythropoiesis in anemia of chronic inflammation (ACI), we previously showed that iron restriction sensitizes cultured human erythroblasts to the inhibitory effects of inflammatory cytokines including interferon γ (IFNγ) (Richardson et al., ASH 2010). This sensitizing effect was reversed by addition of isocitrate to cultures, and in a rat arthritis ACI model intraperitoneal injections of isocitrate completely and durably reversed anemia through in vivo stimulation of erythropoiesis (Richardson et al, ASH 2011). New studies using cultures of human hematopoietic progenitor cells (huHPC) have explored the signaling mechanisms by which iron and isocitrate modulate erythroid responsiveness to IFNγ. No impact of iron restriction or isocitrate treatment could be seen on IFNγ activation of STAT1 phosphorylation on tyrosine 701 or on serine 727. Similarly, iron restriction and isocitrate had no effects on IFNγ-mediated upregulation of STAT1 protein, STAT2 protein, IRF8 protein, or IRF9 mRNA levels. These findings suggest that iron and isocitrate do not affect the classical JAK1-STAT1-IRF1 or the alternative GATE-IRF9 pathways. We then examined expression of the transcription factor PU.1, a master regulator whose levels dictate myeloid versus erythroid cell fate in hematopoietic progenitors. Libregts et al., recently demonstrated that IFNγ upregulated PU.1 erythroblasts via IRF1 (Blood 2011;118(9):2578–2588). Our results showed that iron restriction potently augmented IFNγ induction of PU.1, by 2–3-fold, and also induced PU.1 on its own to a lesser degree. Importantly, isocitrate abrogated the upregulation of PU.1 caused by iron restriction. Furthermore, qRT-PCR on sorted erythroblasts from rat marrows showed increased PU.1 expression in animals with ACI and normalization of erythroid PU.1 expression in association with isocitrate treatment. Pop et al., have recently shown that downregulation of PU.1 early in erythropoiesis constitutes a key step in lineage commitment (PLoS Biol 2010;8(9):e1000484). Therefore we examined the kinetics of PU.1 expression in the huHPC model system. As expected, huHPC downregulated PU.1 during the initial 2–4 days of standard erythroid culture. Similar downregulation occurred in the presence of IFNγ, under iron replete conditions. However, with the combination of IFNγ and iron restriction, PU.1 levels remained high the entire culture period and showed minimal downregulation. Several experimental approaches addressed the erythroid developmental stages affected by iron restriction and IFNγ. Flow cytometry with intracellular staining showed that iron and isocitrate influenced IFNγ induction of PU.1 at an early CD34+ CD36+ stage. These findings were corroborated by immunoblot analysis of sorted progenitors showing iron restriction and isocitrate to affect PU.1 levels within CD36+ GPA- erythroid progenitors; the later CD36+ GPA+ progenitors showed extinction of PU.1 expression regardless of culture conditions. Finally, using purified CD36+ cells as a starting population, iron restriction and IFNγ again cooperated in induction of PU.1, with isocitrate reversing this effect. Prior studies from our lab have shown that erythroid iron restriction results in hyperactivation of PKCα/β. In addition PKCα/β is known to directly phosphorylates and activates PU.1. We therefore sought to determine whether PKC contributes to cooperative upregulation of PU.1 in erythroid progenitors subjected to iron restriction and IFNγ. Supporting this notion, the pan-PKC inhibitor BIM abrogated the effects of iron restriction plus IFNγ on PU.1 upregulation. Importantly, the dosage of BIM employed had no effect on viability or differentiation. Our findings thus define a pathway in which iron restriction and IFNγ act in a cooperative manner on early erythroid progenitors to increase PU.1 expression and interfere with its normal downregulation. Iron restriction and isocitrate exert their influences, at least in part, through alteration of PKC activation. We propose a model of ACI in which iron restriction and inflammatory signaling are both required to attain a critical threshold of erythroid PU.1, which may then interfere with early stages of lineage commitment. Through its reversal of PKC activation by iron restriction, isocitrate may act to keep PU.1 levels below this critical threshold. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 982-982
Author(s):  
Tohru Fujiwara ◽  
Haruka Saitoh ◽  
Yoko Okitsu ◽  
Noriko Fukuhara ◽  
Yasushi Onishi ◽  
...  

Abstract Abstract 982 Background. EZH2, a core component of Polycomb repressive complex 2 (PRC2), plays a role in transcriptional repression through mediating trimethylation of histone H3 at lysine 27 (H3K27), and is involved in various biological processes, including hematopoiesis. Overexpression of EZH2 has been identified in a wide range of solid tumors as well as hematological malignancies. Recent studies indicated that 3-deazaneplanocin A (DZNep), an inhibitor of EZH2, preferentially induces apoptosis in cancer cells, including acute myeloid leukemia and myelodysplastic syndromes, implying that EZH2 may be a potential new target for epigenetic treatment. On the other hand, whereas PRC2 complex has been reported to participate in epigenetic silencing of a subset of GATA-1 target genes during erythroid differentiation (Yu et al. Mol Cell 2009; Ross et al. MCB 2012), the impact of DZNep on erythropoiesis has not been evaluated. Method. The K562 erythroid cell line was used for the analysis. The cells were treated with DZNep at doses of 0.2 and 1 microM for 72 h. Quantitative ChIP analysis was performed using antibodies to acetylated H3K9 and GATA-1 (Abcam). siRNA-mediated knockdown of EZH2 was conducted using Amaxa nucleofection technology™ (Amaxa Inc.). For transcription profiling, SurePrint G3 Human GE 8 × 60K (Agilent) and Human Oligo chip 25K (Toray) were used for DZNep-treated and EZH2 knockdown K562 cells, respectively. Gene Ontology was analyzed using the DAVID Bioinformatics Program (http://david.abcc.ncifcrf.gov/). Results. We first confirmed that DZNep treatment decreased EZH2 protein expression without significantly affecting EZH2 mRNA levels, suggesting that EZH2 was inhibited at the posttranscriptional level. We also confirmed that DZNep treatment significantly inhibited cell growth. Interestingly, the treatment significantly induced erythroid differentiation of K562 cells, as determined by benzidine staining. Transcriptional profiling with untreated and DZNep-treated K562 cells (1 microM) revealed that 789 and 698 genes were upregulated and downregulated (> 2-fold), respectively. The DZNep-induced gene ensemble included prototypical GATA-1 targets, such as SLC4A1, EPB42, ALAS2, HBA, HBG, and HBB. Concomitantly, DZNep treatment at both 0.2 and 1 microM upregulated GATA-1 protein level as determined by Western blotting, whereas the effect on its mRNA levels was weak (1.02- and 1.43-fold induction with 0.2 and 1 microM DZNep treatment, P = 0.73 and 0.026, respectively). Furthermore, analysis using cycloheximide treatment, which blocks protein synthesis, indicated that DZNep treatment could prolong the half-life of GATA-1 protein, suggesting that DZNep may stabilize GATA-1 protein, possibly by affecting proteolytic pathways. Quantitative ChIP analysis confirmed significantly increased GATA-1 occupancy as well as increased acetylated H3K9 levels at the regulatory regions of these target genes. Next, to examine whether the observed results of DZNep treatment were due to the direct inhibition of EZH2 or hitherto unrecognized effects of the compound, we conducted siRNA-mediated transient knockdown of EZH2 in K562 cells. Quantitative RT-PCR analysis demonstrated that siRNA-mediated EZH2 knockdown had no significant effect on the expression of GATA-1 as well as erythroid-lineage related genes. Furthermore, transcription profiles of the genes in the quantitative range of the array were quite similar between control and EZH2 siRNA-treated K562 cells, with a correlation efficient of 0.977. Based on our profiling results, we are currently exploring the molecular mechanisms by which DZNep promotes erythroid differentiation of K562 cells. Conclusion. DZNep promotes erythroid differentiation of K562 cells, presumably through a mechanism not directly related to EZH2 inhibition. Our microarray analysis of DZNep-treated K562 cells may provide a better understanding of the mechanism of action of DZNep. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1543-1543
Author(s):  
Yanfei Xu ◽  
Tanushri Sengupta ◽  
Alexander C. Minella

Abstract Abstract 1543 A growing body of evidence highlights the importance of microRNAs in regulating the expression of mediators of cell cycle progression. A theme emerging from these studies is that microRNAs participate in feedback or feed-forward circuits to provide bistability for key transition points in the cell cycle. We previously have shown that proper regulation of cyclin E activity is required for normal erythroid cell maturation in vivo, using cyclin ET74AüT393A knock-in mice, which have markedly dysregulated cyclin E due to its failure to interact with the Fbw7 ubiquitin ligase complex. We hypothesized that we could identify novel, microRNA-based molecular circuitry for maintaining appropriate levels of cyclin E activity by screening cyclin E knock-in erythroblasts for alterations in microRNA expression. We analyzed data we obtained from multiplex real-time PCR arrays comparing the expression of over 500 microRNAs in cyclin ET74A T393A knock-in versus wild-type erythroblasts (Ter119+/CD71+) and found down-regulated expression of a number of microRNAs targeting CDK inhibitors. We also identified down-regulated expression of potential microRNA regulators of Fbw7 expression. We found that overexpression of miR-223, in particular, significantly reduces Fbw7 mRNA levels, increases endogenous cyclin E protein and activity levels, and increases genomic instability. We next confirmed that miR-223 targets the Fbw7 3’ untranslated region. We then found that reduced miR-223 expression leads to increased Fbw7 expression and decreased cyclin E activity. Finally, we found that miR-223 expression in K562 cells is responsive to acute alterations in cyclin E regulation by the Fbw7 pathway and that dysregulated Fbw7 expression alters the erythroid differentiation capacity of these cells. Mir-223 plays an important role in myeloid and erythroid differentiation by regulating multiple substrates involved in these maturation programs. Here, we identify Fbw7 as a novel target of miR-223. Our data also indicate that miR-223 modulates Fbw7 expression as part of a homeostatic mechanism to regulate cyclin E activity and provide the first evidence that activity of the SCFFbw7 ubiquitin ligase can be controlled by the microRNA pathway. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3388-3388 ◽  
Author(s):  
Felipe R Lorenzo ◽  
Sabina Swierczek ◽  
Chad Daniel Huff ◽  
Josef T. Prchal

Abstract Abstract 3388 The hypoxic response, mediated by hypoxia inducible transcription factors (HIFs), is central to the control and development of many essential biological functions, including erythropoiesis. As a high-altitude population, many Tibetans have developed a remarkable ability to protect against several hypoxic complications, including polycythemia and other harmful responses exhibited by non-adapted populations upon exposure to severe hypoxia. We have identified 10 genes involved in high-altitude adaptation in Tibetans, including a principal negative regulator of HIF-1a and HIF-2 a peptides, i.e. PHD2 (EGLN1), as well as HIF2A (EPAS1) (Simonson, Science 2010). At this meeting last year (Lorenzo, Abstract# 2602 ASH 2010), we reported a novel PHD2 Asp4Glu mutation that we found in 57 of 94 Tibetan, 2 of 88 Asian and 0 of 38 Caucasian chromosomes. In most Tibetan samples, this variant is associated with a previously reported, unvalidated PHD2 polymorphism, Cys127Ser (found in 70 of 94 Tibetan, 27 of 88 Asian and 4 of 38 Caucasian chromosomes). To study the functional consequences of this PHD2 Asp4Glu mutation, we recruited five Tibetan volunteers living in Utah, four of whom were homozygous and 1 heterozygous for PHD2 Asp4Glu and Cys127Ser. We unexpectedly found that homozygotes for the exon 1 PHD2 mutation had markedly hypersensitive erythroid BFU-E (Fig.1) compared to the range of normal controls we have standardized over several decades. Interestingly, erythroid progenitors from individuals with Chuvash polycythemia or a HIF-2a gain-of-function mutation also have hypersensitive BFU-E. To determine if the Tibetan erythroid hypersensitivity data may be explained by increased HIF activity, we have quantified HIF target gene expression in subject granulocytes and found a significant increase in hexokinase (HK1) and glucose transporter (GLUT1/SLC2A) mRNA levels. These data report the first molecular defect with functional consequences that is associated with the complex Tibetan adaptation to extreme hypoxia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3581-3581
Author(s):  
Craig M Forester ◽  
Zhen Shi ◽  
Maria Barna ◽  
Davide Ruggero

Abstract Erythropoiesis constitutes the largest demand on the hematopoietic system due to its extraordinary production on a daily basis. The erythroid proteome requires an integration of multiple external cues to coordinate programs of differentiation as well as maintenance of erythroid precursors. The biomedical relevance of this critical process is underscored by recent findings showing impaired ribosome function in an entire class of clinical disorders with severe impairments in erythroid differentiation, known as ribosomopathies, which remain poorly understood. One of the main signaling pathways controlling post-transcriptional gene expression during erythropoiesis is the mTOR pathway. mTOR activation downstream of SCF/Epo in erythroid progenitors controls the activity of the major cap-binding protein eIF4E. However, the functional role of eIF4E during erythropoiesis and protein synthesis control in this cell type remains unexplored. Here we show that eIF4E activity, through mTOR-dependent phosphorylation of its inhibitory protein 4EBP1, unexpectedly undergoes a dynamic switch between early erythroid precursor populations and during terminal erythrocyte maturation, where eIF4E becomes progressively silenced. Employing a unique eIF4E transgenic mouse model, we strikingly show that overexpression of eIF4E in the bone marrow compartment results in an early accumulation of erythrocyte precursors and a block in erythrocyte differentiation. Surprisingly, this new role of eIF4E in erythropoiesis is independent from control of global protein synthesis but instead may promote a specialized program of translation control that is customized for erythroid cell function. Employing state of the art unbiased proteomics, our work is uncovering distinct networks of proteins, whose expression levels are controlled by eIF4E dosage during specific phases of erythrocyte maturation. Together, our research highlights a novel molecular program linking exquisite regulation of eIF4E activity to specialized translational control underlying erythroid development, providing unprecedented insight into the etiology of erythroid dysfunction in ribosomopathies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 159-159
Author(s):  
Chante Richardson ◽  
Grant C. Bullock ◽  
Lorrie L Delehanty ◽  
Anne-Laure Talbot ◽  
Kamaleldin E Elagib ◽  
...  

Abstract Abstract 159 The anemias of chronic disease (ACD) are a common complication of malignancy, inflammation and kidney disorders. In ACD, there is dysregulation of iron homeostasis, decreased proliferation of erythroid progenitors, diminished production of erythropoietin (EPO), and shortened lifespan of RBC. Multiple pathophysiologic mechanisms have been implicated in the development of ACD, including elevated production of hepcidin and inflammatory cytokines, IFNγ, TRAIL, Interleukins-1β, 6, 10, 15, & TNFα. These cytokines are thought to directly inhibit erythroid differentiation through unknown mechanisms. The current study addressed the hypothesis that inhibition of erythropoiesis in ACD may arise through synergistic effects of iron deprivation and specific inflammatory cytokines. To identify relevant cytokines, candidate factors were applied to primary human erythroid progenitors in iron replete and restricted cultures. Peripheral blood human CD34+ progenitors from healthy donors underwent standard prestimulation for 72 hours, followed by culture in unilineage erythroid medium (4.5 U/ml EPO + 25ng/ml SCF) for 4-5 days under iron replete (100% transferrin saturation) or iron restricted (15% transferrin saturation) conditions. Candidate cytokines were screened for effects on viability, proliferation, and differentiation using cell counting and flow cytometric analysis of the erythroid cell surface marker GPA and the megakaryocytic antigen CD41a. Contrary to previous reports, the majority of cytokines (TRAIL & Interleukins-1β, 6, 10, 15) showed no effects on erythroid proliferation or differentiation under iron replete or restricted conditions. By contrast, both IFNγ and TNFα displayed potent inhibitory effects under iron restricted conditions but only weak effects in iron replete cultures. Typically, iron restriction alone reduced the proportion of GPA+ cells by 50%, whereas IFNγ or TNFα combined with iron restriction caused a 90% reduction. While both cytokines cooperated with iron restriction in blocking upregulation of GPA and promoting cell death, each cytokine also had distinctive effects on morphology and differentiation. IFNγ enhanced megakaryocytic development, while TNFα retained cells as immature, CD34+ progenitors. The synergistic inhibition of erythroid differentiation with iron restriction and TNFα was confirmed in vivo using a murine model of dietary iron deprivation coupled with continuous infusion of low-dose TNFα. Regarding the mechanism for this synergy, we have previously shown that erythroid iron deprivation leads to inactivation of the aconitase enzymes, which normally convert citrate to isocitrate, and that provision of exogenous isocitrate abrogates the erythroid inhibition associated with iron deprivation. Accordingly, participation of this pathway was assessed in the more potent erythroid inhibition associated with IFNγ or TNFα plus iron deprivation. Strikingly, isocitrate administration not only abrogated effects due to iron deprivation but also those due to the inflammatory cytokines, leading to complete rescue of erythroid differentiation. To address the underlying basis for erythroid cross-talk of iron and cytokine signaling, we screened pathways implicated in iron metabolism and inflammation. Two relevant pathways identified were Jun kinase (JNK) and calmodulin-associated kinase II (CAMKII), important in TNFα and IFNγ signaling, respectively. In particular, TNFα and iron deprivation synergized in the activation of JNK, and IFNγ and iron deprivation synergized in activating CAMKII. In both cases, isocitrate partially restored the activation to basal levels. As an important negative control, iron deprivation did not affect IFNγ activation of STAT1 phosphorylation, indicating that its effects were not due to upregulation of receptor expression or function. Altogether, these data suggest that among the various cytokines implicated in ACD, only IFNγ and TNFα synergize with iron deprivation in the inhibition of erythropoiesis. These actions occur through cross-talk between intracellular signaling pathways, specifically pathways involving aconitase and cytokine-activated kinases. The connection of aconitase/metabolism with inflammation is novel and has implications for clinical treatment of ACD, as well as for new understanding of erythroid and inflammatory signaling. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4238-4238
Author(s):  
Seema Singh ◽  
Arvind Dev ◽  
Pradeep Sathyanarayana ◽  
Donald J McCrann ◽  
Christine Emerson ◽  
...  

Abstract Abstract 4238 In late stage erythroblasts, EPO can increase levels of Bclx, Bcl2 and/or Mcl1 anti-apoptotic factors. Proerythroblasts, however, are a key EPO target (and exhibit sharp dependence on EPO for growth, and survival). In these progenitors, however, Bclx, Bcl2 and Mcl1 are not prime EPO/EPOR targets. Via transcriptome-based analyses of EPO response circuits in developmentally staged primary bone marrow proerythroblasts (which we now analyze and present at a global level) an atypical TNF receptor, Tnfrsf13c proved to be among the top 1% of EPO/EPOR induced factors. Within lymphoid lineages, Tnfrsf13c is a known receptor for BAFF ligand; and BAFF is an essential mediator of B-cell survival and development. Possible effects of BAFF (a bone marrow stromal cell surface ligand) on primary erythroid cell formation therefore were assessed. Notably, limited BAFF exposure (15 hours) inhibited apoptosis; increased erythroid cell numbers; and enhanced the formation of late-stage Ter119pos erythroblasts. Specifically, cytoprotection by BAFF rivaled that afforded by EPO; cell numbers were enhanced 140% (in 15 hr); and frequencies of Ter119pos erythroblasts were enhanced to 200% of controls. In keeping with Tnfrsf13c's role as an EPOR target, each of the above effects further proved to depend upon proerythroblast exposure to EPO. With regards to Tnfrsf13c expression, analyses using primary erythroid progenitors with knocked-in minimal EPOR alleles indicated dependence for EPO- induction upon JAK2, STAT5 as well as EPOR C-terminal coupled pathways. Studies overall reveal a novel EPOR action route within primary proerythroblasts as a Tnfrsf13c/BAFF pathway (which engages non-canonical NF-kappaB molecular mechanisms). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4172-4172
Author(s):  
Richard XuFeng ◽  
Moo-Rim Park ◽  
Weimin Miao ◽  
Hui Yu ◽  
Michael W. Epperly ◽  
...  

Abstract Abstract 4172 A tightly regulated level of reactive oxygen species (ROS) is critical for proper functioning of hematopoietic stem cells (HSCs) and high levels of ROS were shown to be able to exhaust the HSC compartment under stress conditions. Intracellular excessive ROS are normally scavenged by antioxidant enzymes. Superoxide anion radical (O) converts into hydrogen peroxide (H2O2) by superoxide dismutase (SOD), and then detoxified by catalase and glutathione peroxidase (GPx) into water. We hypothesize that maintaining a low redox status by ectopically expressing these ROS-detoxifying enzymes, namely manganese-containing SOD (MnSOD) and catalse is able to augment HSC regeneration. In this study, we explored the potential usefulness of MnSOD-PL, a drugable MnSOD plasmid and lipfectin complex, in enhancing the efficiency of HSC transplant. We have also investigated the effects of overexprressed MnSOD or catalase via retroviral delivery on the engraftment efficiency of transduced HSCsSCs. Our results showed that the basal mRNA levels of MnSOD and catalase in long-term and short-term HSCs were less than 1–3% of that of the cellular beta-actin. The mRNA levels were 2 to 4 fold higher in the short-term repopulating HSCs than those in the long-term repopulating HSCs. Irradiation did not induce enzymes expression except catalase in megakaryocytic-erythroid progenitors (MEP) and GPx1 in granulocyte-monocytic progenitors (GMP), which were significantly increased after exposure to 800 cGy (p=0.05) and 400 cGy (p=0.046) radiations, respectively. Administration of MnSOD-PL before TBI conferred significant radiation protection for irradiated recipient mice. On the one hand, the recovery of endogenous hematopoietic could be boosted. On the other hand, the function of the donor HSCs was more preserved from the ROS damage and proliferative stress, and those preserved HSCs were able to replenish the BM when a secondary stress occurred. However, the beneficial effects of MnSOD-PL seemed to be largely via the host environment, since our subsequent experiment showed that overexpression of MnSOD in HSCs only provided minimal benefits to the hematopoietic reconstitution. This potential utility of MnSOD-PL suggests an alternative therapeutic strategy to enhance the HSCs engraftment efficiency in bone marrow transplant. In order to further explore the effects of catalase in HSC protection, human catalase was over-expressed in mouse LKS cells via retroviral delivery in comparison with MnSOD. In the colony-forming cell assay, CFU-M colonies were significantly higher in catalase or MnSOD over-expressed group than those of vector control group in the 200 cGy irradiated plates (p=0.01). Within three months after competitive transplantation, the engraftment levels were increased to 2.7–3.4 fold in catalase group than those of vector group. After 200 cGy re-irradiation engraftment levels were significantly increased to 6.4–7.9 fold in the catalase group (p<0.05). The results showed that overexpression of catalase alone could significantly improve the HSC repopulation. Despite no significant difference in the primary transplantation experiments, MnSOD over-expression also showed a positive effect on the HSC repopulation capacity in the serial transplantation. Taken together, these results demonstrate that maintaining low ROS by ectopically expressing the ROS-detoxifying enzymes is a viable approach to improve HSC functions under stress and damaging conditions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1175-1175
Author(s):  
Vikram R Paralkar ◽  
Cristian Taborda ◽  
Yu Yao ◽  
Rishi Prasad ◽  
Jing Luan ◽  
...  

Abstract Mammalian cells express thousands of lineage-restricted long noncoding RNAs (lncRNAs), although the functions of most are unknown. "Knockout" studies to investigate lncRNA function by gene deletion in cells or whole animals have been informative. However, since most lncRNAs show open chromatin and transcription factor (TF) binding at their promoters as well as elsewhere in their genomic loci, it is essential to distinguish the roles of the RNA transcript from those of potential cis-regulatory elements, both of which are eliminated in deletion studies. We identified a novel mouse lncRNA named Drop27 (Downstream of p27) that is transcribed 4 kb downstream of the Cdkn1b gene encoding the cell-cycle inhibitor p27, which is strongly upregulated during erythroid maturation. Drop27 is conserved between mice and humans, has two exons separated by a 21 kb intron, and produces a 400 bp spliced, polyadenylated RNA that is abundantly transcribed and highly enriched in mouse erythroblasts compared to other tissues. We used the CRISPR/Cas system to delete the entire Drop27 locus in the G1E erythroid cell line. Microarray analysis showed reduced Cdkn1b mRNA in Drop27-deleted cells with very few other changes in the transcriptome. Quantitative PCR verified this finding by demonstrating that heterozygous deletion of Drop27 caused reduced Cdkn1b mRNA by 35%, while homozygous deletion led to a 70% reduction. This effect also occurred at the primary transcript level, demonstrating that deletion of the Drop27 reduced Cdkn1b transcription. To distinguish the effects of loss of the lncRNA transcribed from Drop27 from the potential loss of a cis-acting regulatory module in the Drop27 genomic locus, we used homologous recombination to insert a bovine growth hormone (BGH) polyadenylation (polyA) cassette into the first exon of Drop27, 80 bp downstream of the transcription start site. This caused premature termination of the lncRNA transcript while maintaining all genomic sequences, including potential cis-regulatory elements. Analysis of multiple clones showed that homozygous polyA cassette integration reduced full-length Drop27 transcript levels by more than 99%, although Cdkn1b mRNA levels were unaffected. These findings demonstrate that Drop27 lncRNA is dispensable for Cdkn1b transcription, while its genomic sequences are required, indicating that the Drop27 gene locus contains an erythroid cis-regulatory element (enhancer) for the Cdkn1b gene. Several strong candidates for the proposed enhancer are found in the Drop27 gene. Multiple epigenetic features strongly associated with enhancers map in several distinct locations in the Drop27 locus in erythroid and other hematopoietic cells, including DNase hypersensitivity, p300 binding, and multiple transcription factor sites. A functional role for the Drop27 lncRNA is not identified by this experiment, and it is possible that it arose as a byproduct of enhancer activity. Our findings provide new insight into mechanisms of hematopoietic gene expression and are of more broad relevance to the lncRNA field in general. In particular, we demonstrate that the genomic loci of some lncRNA genes may function as cis elements, irrespective of the transcripts arising from them. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3203-3203
Author(s):  
Saswati Sen ◽  
Hanming Wang ◽  
Sally-lin Adams ◽  
Janice Yau ◽  
Kim Zhou ◽  
...  

Abstract Abstract 3203 Poster Board III-140 Anemia occurs in 60% of patients with Shwachman Diamond Syndrome (SDS). Although bi-allelic mutations in SBDS cause SDS, it is unclear whether SBDS is critical for erythropoiesis and what the pathogenesis of anemia is in SDS. We hypothesize that SBDS protects early erythroid progenitors from apoptosis by promoting ribosome biosynthesis and translation. During early erythroid differentiation of human K562 cells and primary CD133+ cells, a prominent upregulation of SBDS by RT-qPCR was found. SBDS deficiency by vector-based shRNA led to impaired cell expansion of differentiating K562 cells due to accelerated apoptosis and a mild reduction in proliferation. Furthermore, the cells showed general reduction of 40S, 60S, 80S ribosomal subunits, loss of polysomes and impaired global translation during differentiation. Both cell expansion and translation defects were rescued upon re-introduction of SBDS in K562 cells. Interestingly, leucine partly corrected the cell expansion and translational defects of non-differentiating SBDS-deficient K562 cells, while differentiating SBDS-deficient K562 cells showed improved cell expansion in the presence of additional translation stimulators such as IGF-1. SBDS-knockdown CD133+ cells showed increased BFU-E colony formation under conditions with leucine and a combination of leucine and IGF-1 treatment. Although the erythroid cell expansion defect in K562 cells is independent of p53 as these cells do not express the gene, an upregulation of TAp73, was found in resting SBDS deficient K562 cells. However expression of TAp73 was lost during differentiation. DNp63 was also not upregulated in SBDS-deficient K562 erythroid cells. These results demonstrate that the role of SBDS in non-differentiated cells versus differentiated cells represents two dynamic scenarios and that SBDS plays a critical role in erythroid expansion by promoting survival of early erythroid progenitors and in maintaining ribosome biogenesis during erythroid maturation through a pathway independent of p53 family members. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1336-1336
Author(s):  
Song Jihyun ◽  
Yoon Donghoon ◽  
Robert D. Christensen ◽  
Perumal Thiagarajan ◽  
Josef T. Prchal

Abstract Oxygen homeostasis is tightly controlled by the number of red blood cells (RBCs). Hypoxia increases RBCs by enhanced erythropoiesis mediated by hypoxia-inducible factors (HIFs). Upon return to normoxia, the increase in RBCs is overcorrected by preferential destruction of young RBCs, a process termed neocytolysis. Neocytolysis was first described in astronauts and later in people descending from high altitude. The molecular mechanism of neocytolysis is obscure. We hypothesized that neocytolysis occurs because of rapid transient changes of HIF levels, resulting in increased reactive oxygen species (ROS) from mitochondria in reticulocytes and defective antioxidant protection of young RBCs generated in hypoxia. We developed a neocytolysis model by exposing mice to 12% oxygen (equivalent to 4500m altitude) for 10 days. This model recapitulates the RBC changes observed in humans exposed to hypoxia for ~30 days. Upon return to normoxia, ROS were markedly increased in reticulocytes and mature RBCs, but not in neutrophils, B- or T-cells, or monocytes. Reduction of ROS by antioxidant (N-acetyl-L cysteine) treatment attenuated hemolysis and decreased hematocrit. To test whether HIFs (transcription factors regulating hypoxic response) contribute to neocytolysis, we repeated these experiments using Chuvash mice (which bear a VHLR200Wmutation, resulting in constitutively high HIF). These mice also showed attenuated hemolysis and decreased hematocrit; in addition, their reticulocyte half-life was higher (36.6 vs.17.8 hours in wild type). Similar findings were also observed with treatment of mice with dimethyloxallyl glycine (DMOG), an inhibitor of prolyl hydroxylase (another negative regulator of HIF). These experiments indicate the essential role of HIF pathways in neocytolysis. Mitochondria are a major source of ROS in cells. During terminal differentiation of RBCs, mitochondria are removed from reticulocytes by mitophagy. After hypoxia treatment, mitochondria mass increased in reticulocytes concomitant with the reduction of HIF-regulated Bnip3L (a mediator of mitophagy) transcripts. These increased ROS were of mitochondrial origin, as detected by Mito-Sox staining. To pursue the mechanism behind the preferential destruction of young RBCs, we investigated antioxidant enzymes. After hypoxia treatment, catalase decreased by 30%, but not glutathione peroxidase, superoxide dismutase or NADH oxidase. The decreased catalase in RBCs produced during hypoxia was unexpected, as it was shown previously that catalase is regulated by HIF2 (as we also show, to a lower degree by HIF1 in our Hif1a-/-embryo) suggesting alternate negative regulator(s) of catalase in hypoxia. Several hypoxia-regulated microRNAs (miRs) are reported to control oxidative stress; we found that miR-451, miR-205 and miR-21 were expressed in erythroid progenitors and reticulocytes and induced after 10 day-hypoxia exposure. To verify whether these miRs regulate catalase expression, we overexpressed and downregulated these miRs in K562 and HEL erythroid cell lines, and found that only miR-21 regulated catalase. Further, we found increased miR-21 after 10 day-hypoxia exposure, with a concomitant decrease of catalase transcripts and activity resulting in impaired ROS scavenging. We conclude that neocytolysis is mediated by excessive generation of ROS from increased mitochondrial mass due to reduced Bnip3L in reticulocytes upon return to normoxia. The reticulocyte ROS then interact with hypoxia-produced young RBCs having miR-21-downregulated catalase, resulting in their preferential destruction. We show that increased mitochondrial ROS and miR-21-downregulated catalase provide the molecular basis of neocytolysis. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document