scholarly journals Decreased PGC1β expression results in disrupted human erythroid differentiation, impaired hemoglobinization and cell cycle exit

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Taha Sen ◽  
Jun Chen ◽  
Sofie Singbrant

AbstractProduction of red blood cells relies on proper mitochondrial function, both for their increased energy demands during differentiation and for proper heme and iron homeostasis. Mutations in genes regulating mitochondrial function have been reported in patients with anemia, yet their pathophysiological role often remains unclear. PGC1β is a critical coactivator of mitochondrial biogenesis, with increased expression during terminal erythroid differentiation. The role of PGC1β has however mainly been studied in skeletal muscle, adipose and hepatic tissues, and its function in erythropoiesis remains largely unknown. Here we show that perturbed PGC1β expression in human hematopoietic stem/progenitor cells from both bone marrow and cord blood results in impaired formation of early erythroid progenitors and delayed terminal erythroid differentiation in vitro, with accumulations of polychromatic erythroblasts, similar to MDS-related refractory anemia. Reduced levels of PGC1β resulted in deregulated expression of iron, heme and globin related genes in polychromatic erythroblasts, and reduced hemoglobin content in the more mature bone marrow derived reticulocytes. Furthermore, PGC1β knock-down resulted in disturbed cell cycle exit with accumulation of erythroblasts in S-phase and enhanced expression of G1-S regulating genes, with smaller reticulocytes as a result. Taken together, we demonstrate that PGC1β is directly involved in production of hemoglobin and regulation of G1-S transition and is ultimately required for proper terminal erythroid differentiation.

Leukemia ◽  
2014 ◽  
Vol 29 (5) ◽  
pp. 1092-1103 ◽  
Author(s):  
H Dolatshad ◽  
A Pellagatti ◽  
M Fernandez-Mercado ◽  
B H Yip ◽  
L Malcovati ◽  
...  

Abstract The splicing factor SF3B1 is the most commonly mutated gene in the myelodysplastic syndrome (MDS), particularly in patients with refractory anemia with ring sideroblasts (RARS). We investigated the functional effects of SF3B1 disruption in myeloid cell lines: SF3B1 knockdown resulted in growth inhibition, cell cycle arrest and impaired erythroid differentiation and deregulation of many genes and pathways, including cell cycle regulation and RNA processing. MDS is a disorder of the hematopoietic stem cell and we thus studied the transcriptome of CD34+ cells from MDS patients with SF3B1 mutations using RNA sequencing. Genes significantly differentially expressed at the transcript and/or exon level in SF3B1 mutant compared with wild-type cases include genes that are involved in MDS pathogenesis (ASXL1 and CBL), iron homeostasis and mitochondrial metabolism (ALAS2, ABCB7 and SLC25A37) and RNA splicing/processing (PRPF8 and HNRNPD). Many genes regulated by a DNA damage-induced BRCA1–BCLAF1–SF3B1 protein complex showed differential expression/splicing in SF3B1 mutant cases. This is the first study to determine the target genes of SF3B1 mutation in MDS CD34+ cells. Our data indicate that SF3B1 has a critical role in MDS by affecting the expression and splicing of genes involved in specific cellular processes/pathways, many of which are relevant to the known RARS pathophysiology, suggesting a causal link.


Blood ◽  
2006 ◽  
Vol 107 (11) ◽  
pp. 4291-4299 ◽  
Author(s):  
Binghui Li ◽  
Nan Jia ◽  
Reuben Kapur ◽  
Kristin T. Chun

AbstractAs erythroid progenitors differentiate into precursors and finally mature red blood cells, lineage-specific genes are induced, and proliferation declines until cell cycle exit. Cul4A encodes a core subunit of a ubiquitin ligase that targets proteins for ubiquitin-mediated degradation, and Cul4A-haploinsufficient mice display hematopoietic dysregulation with fewer multipotential and erythroid-committed progenitors. In this study, stress induced by 5-fluorouracil or phenylhydrazine revealed a delay in the recovery of erythroid progenitors, early precursors, and normal hematocrits in Cul4A+/– mice. Conversely, overexpression of Cul4A in a growth factor-dependent, proerythroblast cell line increased proliferation and the proportion of cells in S phase. When these proerythroblasts were induced to terminally differentiate, endogenous Cul4A protein expression declined 3.6-fold. Its enforced expression interfered with erythrocyte maturation and cell cycle exit and, instead, promoted proliferation. Furthermore, p27 normally accumulates during erythroid terminal differentiation, but Cul4A-enforced expression destabilized p27 and attenuated its accumulation. Cul4A and p27 proteins coimmunoprecipitate, indicating that a Cul4A ubiquitin ligase targets p27 for degradation. These findings indicate that a Cul4A ubiquitin ligase positively regulates proliferation by targeting p27 for degradation and that Cul4A down-regulation during terminal erythroid differentiation allows p27 to accumulate and signal cell cycle exit.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3796-3796 ◽  
Author(s):  
Rajasekhar NVS Suragani ◽  
Robert Li ◽  
Dianne Sako ◽  
Asya Grinberg ◽  
R. Scott Pearsall ◽  
...  

Abstract Abstract 3796 Myelodysplastic syndromes (MDS) are a group of hematopoietic stem cell disorders characterized by peripheral blood cytopenias such as anemia, neutropenia or thrombocytopenia. Ineffective erythropoiesis due to increased proliferation and abortive maturation of precursors leads to severe anemia, the most common cytopenia observed in MDS syndromes. Despite elevated erythropoietin (EPO) and erythroid hyperplasia, MDS patients are often given recombinant EPO therapy to stimulate erythropoiesis. However, only a small proportion of patients respond to EPO therapy. Frequent blood transfusions as supportive care result in iron overloading and recently iron overloading is also linked to enhanced progression to AML. Therefore, alternative therapies are necessary to treat anemia in MDS patients. Signaling by members of the TGFβ superfamily are known regulators of erythropoiesis. We developed ACE-536, a ligand trap consisting of a modified activin receptor Type IIB extracellular domain linked to a human Fc domain. In vitro assays revealed that ACE-536 inhibits smad 2/3 ligands of the signaling pathway but not smad 1/5/8 ligands. Dose dependent studies using ACE-536 in mice, rats and monkeys revealed that ACE-536 treatment resulted in increased red blood parameters but did not affect other cell types. These data suggests that ACE-536 inhibits smad 2/3 phosphorylation modulating the expression of downstream genes involved in erythroid development pathway. BFU-E and CFU-E colony formation assays from bone marrow and spleen in mice following ACE-536 treatment revealed that ACE-536 did not affect the proliferation stages of erythropoiesis. In mice, terminal erythroid differentiation analysis by flow cytometry at 72hrs following RAP-536 (10mg/kg) treatment demonstrated decreased basophilic and increased ortho- and poly-chromatophilic erythroblasts and reticulocytes compared to VEH treatment. Cell cycle analysis of bone marrow and splenic erythroblasts counterstained with BrdU and 7-AAD after RAP-536 (10mg/kg, for 24 hours) or VEH treatment to EPO pre-treated (1500 units/kg, for 40 hours) mice (N=5/group) revealed that EPO+RAP-536 treatment resulted in significant decrease in S-phase and increase in G1/G2-phases of cell cycle compared to EPO+VEH treatment. In addition, EPO+RAP-536 treatment resulted in a greater increase in RBC parameters than either of the treatments alone. Together, these results demonstrate that ACE-536 increases red blood cell formation by promoting maturation of late stage erythroblasts. We then investigated the effect of ACE-536 on anemia in NUP98-HOXD13 (NHD13) transgenic murine model of MDS. NHD13 mice develop anemia, neutropenia and lymphopenia, with normal or hyper cellular bone marrow. A Majority of the mice die by 14 months due to severe pancytopenia or progression to acute myeloid leukemia. In this study, mice were divided into three groups based on age. Early (∼4 months old), mid (∼8 months old) and late stage (∼10 months) groups were randomized and dosed with either RAP-536 at 10 mg/kg or VEH twice per week for 6–8 weeks. NHD13 mice in each group had severe anemia characterized by reduced RBC, Hemoglobin and HCT and compared to wild-type littermates prior to treatment. Treatment of RAP-536 for 6–8 weeks significantly increased RBC parameters and reversed anemia at all stages. Peripheral blood smear analysis revealed no indication of increased leukemic progression due to RAP-536 treatment. Cell differential and flow cytometric evaluation of erythroid precursors from bone marrow demonstrated decreased erythroid precursors and hyperplasia after RAP-536 treatment compared to vehicle treated control. Our data demonstrate that RAP-536 can increase hematology parameters by enhancing maturation of terminally differentiated red blood cells. We have shown RAP-536 corrects ineffective erythropoiesis, decreases erythroid hyperplasia and normalizes myeloid: erythroid ratios without enhanced progression to AML in a murine MDS model. Therefore ACE-536 may represent a novel treatment for anemia associated with MDS, particularly in patients that are refractory to EPO therapy. ACE-536 has completed Phase I clinical trials in healthy human volunteers and Phase II study in MDS patients is planned. Disclosures: Suragani: Acceleron Pharma Inc: Employment, Equity Ownership. Li:Acceleron Pharma Inc: Employment, Equity Ownership. Sako:Acceleron Pharma Inc: Employment, Equity Ownership. Grinberg:Acceleron Pharma Inc: Employment, Equity Ownership. Pearsall:Acceleron Pharma Inc: Employment, Equity Ownership. Kumar:Acceleron Pharma Inc: Employment, Equity Ownership.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 697-697
Author(s):  
Xu Han ◽  
Jieying Zhang ◽  
Yuanliang Peng ◽  
Huiyong Chen ◽  
Xiao Chen ◽  
...  

Abstract Erythropoiesis is a process during which hematopoietic stem cell (HSCs) are first committed to erythroid progenitors, which subsequently undergo terminal erythroid differentiation to produce mature red blood cells. During terminal erythroid differentiation, proerythroblasts undergo 4-5 mitoses to sequentially generate basophilic erythroblasts, polychromatic erythroblasts and orthochromatic erythroblasts that expel their nuclei to produce enucleated reticulocytes. Terminal erythropoiesis is a tightly regulated process. The most well studied regulatory mechanisms include EPO/EPOR mediated signal transduction and transcription factors among which GATA1 plays critical role. Terminal erythroid differentiation is also tightly coordinated with cell cycle exit, which is regulated by cyclins, cyclin-dependent kinases and cyclin-dependent kinase inhibitors (CDKI), yet their roles in erythropoiesis remain largely undefined. Our RNA-seq of human terminal erythroid differentiation shows that of seven CDKI members, only three of them, p18INK4c, p19INK4d and p27KIP1, are abundantly expressed in erythroid cells and their expressions are significantly upregulated in late stage erythroblasts, which were further confirmed by western blotting analysis. In contrast to demonstrated roles of p18INK4c and p27KIP1 in terminal erythroid differentiation, the function of p19INK4d this process has not been studied. To explore the role of p19INK4d during human erythropoiesis, we employed a shRNA-mediated knockdown approach in CD34+ cells and found that p19INK4d knockdown delayed erythroid differentiation, inhibited cell growth, led to increased apoptosis and generation of abnormally nucleated late stage erythroblasts. Unexpectedly, p19INK4d knockdown did not affect cell cycle. Rather it led to decreased GATA1 protein levels. Importantly, the differentiation and nucleus defects were rescued by ectopic expression of GATA1. As GATA1 protein is protected by nuclear HSP70, to explore the mechanism for the decreased GATA1 protein levels, we examined the effects of p19INK4d knockdown on HSP70 and found p19INK4d knockdown led to decreased nuclear localization of HSP70 due to reduced ERK activation. Further biochemical analysis revealed that p19INK4d directly binds to Ras kinase inhibitor PEBP1 and that p19INK4d knockdown increased the expression of PEBP1 which in turn led to reduced ERK activation. These results demonstrate that p19INK4d maintains GATA1 protein levels through PEBP1-pERK-HSP70-GATA1 pathway. Our findings identify previously unknown and unexpected roles for p19INK4d in human terminal erythroid differentiation via a novel pathway. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 945-945
Author(s):  
Rialnat Adebisi Lawal ◽  
Kathleen E. McGrath ◽  
Laura M. Calvi

Abstract Osteolineage cells within the bone marrow microenvironment have been implicated in support and regulation of hematopoietic stem cells (HSCs). Recently, augmented hypoxia-inducible factor (HIF) signaling in osteoprogenitors has been shown to expand the HSC niche, and surprisingly these cells have also been demonstrated to express erythropoietin, the critical cytokine stimulating erythropoiesis. We therefore hypothesize that endosteal cells may represent an additional regulatory site for erythropoiesis. To further delineate the role of the osteolineage cells in the support of erythropoiesis, we isolated bone associated cells (BACs) with enzymatic digestion of adult C57bl/6 mice hind limbs after bone marrow flushing and depleted the BACs of CD45+ cells to enrich for osteogenic cells. We suspected some contribution of erythroid cells to CD45- BACs, however we were surprised to find that ter119+ cells represented a large percentage of BACs after enzymatic digestion. After CD45 depletion, ter119+ cells constituted about 30% percent compared to approximately 0.85% of CD45+ cells (33 ± 4.4vs. 0.85 ± 0.26, p= 0.0018) by flow cytometric analysis. Additionally, CD45 depleted BACs had approximately 46 fold higher osteocalcin expression than CD45+ cells (1300 ± 120 vs. 28 ± 9.5, p < 0.0001), while CD45/Ter119/CD31 depleted BACs had approximately 2000 fold higher osteocalcin expression than CD45/Ter119/CD31 (+) cells (2000 ± 520 vs. 0.98 ± 0.02, p= 0.0044) by qRT-PCR, confirming enrichment of the osteoblastic lineage by this immunophenotypic panel. These data suggest that there are a large number of erythroid lineage cells associated with the BACs along the endosteum. In the bone marrow of adult mice, ter119 + cells represented approximately 85% in the CD45- pool as compared to 5% in the CD45+ cell pool. To determine if the endosteum is an active site of erythropoiesis, we quantified erythroid progenitors and precursors in the BAC pool compared to whole bone marrow (wbm) and peripheral blood (pb) by both flow cytometric analysis and colony forming assays. Flow cytometric analysis demonstrated the presence of every phase of erythroid differentiation in the BAC pool, including the presence of phenotypic MEPs (wbm vs bac vs pb: 250 ± 30 vs 84 ± 22 vs 0), BFU-E (wbm vs bac vs pb: 300 ± 14 vs 110 ± 36 vs 0 ), CFU-E (wbm vs bac vs pb: 2900 ± 2 vs 430 ± 23 vs 1 ± 0.8) and proerythroblasts (wbm vs bac vs pb: 11000 ± 2500 vs 7600 ± 1600 vs 2300 ± 920) per million cells. The phenotypic frequency of CFU-E was particularly remarkable in the BACs (430 ± 23) as compared to peripheral blood (1 ± 0.8) , demonstrating that all stages of erythroid differentiation are found in tight association with the endosteum and are not due to contamination from circulating erythroid progenitors. Colony assays were performed for CFU-E (wbm vs. bac 108 ± 16 vs 6.3 ± 2 colonies per 20,000cells plated), BFU-E (wbm vs. bac 55 ±1.0 vs 2 ±1.0; colonies per 40,000 cells plated) and myeloid progenitors (wbm vs. bac 66 ± 28 vs 11 ± 2.5 ; colonies per 10,000 cells plated) also confirmed the presence of erythroid progenitors at endosteal sites. Together these results identify the endosteal surface as a site for erythroid differentiation. The presence of all phases of erythroid lineage differentiation in the BACs suggests a potential role for osteolineage cells for maintenance and regulation of erythropoiesis. Whether osteolineage cells contribute to erythroid lineage homeostasis and/or stress response, and whether activation or damage to osteolineage cells alters local erythroid differentiation remains to be demonstrated. However our data suggest further study of the endosteum and osteolineage cells as a potential and unexpected site of erythroid regulation, which could potentially be targeted to accelerate erythropoiesis and treat anemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 638-638
Author(s):  
Vijay G. Sankaran ◽  
Carl R. Walkley ◽  
Bruce M. Spiegelman ◽  
Stuart H. Orkin

Abstract Regulation of the cell cycle is intimately linked to erythroid differentiation, yet how these processes are coupled at the molecular level is not well understood. In order to gain insight into this coordinate regulation, we examined the role that the retinoblastoma gene product (Rb), a central regulator of the cell cycle, plays in erythropoiesis. A conditional (floxed) Rb gene was specifically deleted in the erythroid compartment by means of EpoR-GFPcre, or in other hematopoietic compartments (with LysM-Cre and Mx1-Cre) in mice. We found that Rb serves a cell-type intrinsic and autonomous role in erythropoiesis. Deletion of Rb in the erythroid compartment results in an anemia due to ineffective erythropoiesis. A partial block in differentiation occurs at the transition from early to late erythroblasts. Unexpectedly, we found that in addition to a failure to properly exit the cell cycle, mitochondrial biogenesis fails to be upregulated at this transition point. The disturbance in mitochondrial activities was established through gene expression analysis, measures of respiratory function, as well as measurements of mtDNA content in sorted cell populations. The failure to upregulate mitochondrial biogenesis appears to contribute to the block in differentiation. The link between erythroid differentiation and mitochondrial function was validated by chemical inhibition of mitochondrial biogenesis and knockdown of the mitochondrial transcriptional coactivator PGC-1beta in cultured erythroid cells. The ineffective erythropoiesis seen in erythroid cells lacking Rb closely resembles the features of the refractory anemia seen in the context of the myelodysplastic syndromes (MDS). Prior work has demonstrated the presence of defects in cell cycle regulation as well as mitochondrial function in MDS. Our work demonstrates how these seemingly disparate pathways may play a role in coordinately regulating cellular differentiation.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1192-1192
Author(s):  
Yoann Vial ◽  
Jeannette Nardelli ◽  
Justine Rousselot ◽  
Emilie Lepeu ◽  
Michele Souyri ◽  
...  

Introduction: Microcephaly is a recurrent feature in patients with inherited bone marrow failure (iBMF) and DNA damage response (DDR) disorders suggesting that common cellular pathways regulate the proliferation and differentiation of neural and hematopoietic progenitors. However, while several studies addressed the role of iBFM or DDR genes in brain development, a possible role for microcephaly genes in hematopoietic development has not been investigated. To address this issue, we studied a mouse model of primary microcephaly with biallelic loss-of-function in Mcph1.MCPH1 mutations are found in 10% of patients with isolated forms of genetic microcephaly (MCPH). Interestingly, MCPH1 helps to maintain genomic integrity during cell division by interacting with proteins involved in cell cycle progression, apoptosis or DNA repair, all cellular processes being also involved in iBMF and DDR syndromes. Methods: Mcph1 null mice were generated by germline deletion of Mcph1 exon 2 (Mcph1tm1.2Kali) (Liang et al., PLoS Genet., 2010). The subpopulations of erythroid progenitors S0 to S5 were phenotypically defined and sorted by flow cytometry according to CD71 and Ter119 expression in the Lin- compartment from mouse liver obtained at birth and during fetal development (E12.5). RNA sequencing (RNA-Seq) was performed on sorted erythroid progenitor fractions obtained from E12.5 fetal livers (SMARTer® Stranded Total RNA-Seq Kit V2-Pico Input library preparation kit). Cell division was studied by multiplexing erythroid specific antibodies with EdU flow cytometry cell proliferation assay. Results: Null mice recapitulated the microcephaly phenotype seen in MCPH patients, but also showed a striking anemic pallor. Numeration and cytomorphologic examination of peripheral blood at birth confirmed macrocytic anemia with low red blood cell count and anisopoikilocytosis. These observations were consistent with congenital dyserythropoiesis in Mcph1-/- mice and prompted us to further characterize the erythroid lineage. Quantification of erythroid progenitor populations in liver at birth showed a significant decreased from the S3 subset (Lin-, CD71High, Ter119High) suggesting impaired terminal differentiation. Similar results were obtained in fetal livers at E12.5 indicating that the defect arose early in hematopoietic ontogeny. Transcriptome analysis of wild-type progenitor populations (S0 to S3) confirmed that Mcph1 is expressed during normal erythropoiesis following a Gata1-like expression profile. This is consistent with the presence in the Mcph1 gene promoter of a binding site for Gata1 and Ldb1 (ENCODE project), supporting an activation by the main erythroid differentiation complex. Strikingly, RNA-Seq analysis revealed deregulation of p53 pathway associated genes in all subsets of Mcph1-/- erythroid progenitors as compared to their wild-type counterparts. Two transcriptional p53 targets involved in cell cycle control, Cdkn1a coding the cyclin-dependent kinase inhibitor (p21) and Ccng1 coding Cyclin G1, were among the most upregulated genes. Cell cycle analysis performed on sorted erythroid progenitors revealed an endoreduplication phenomenon restricted to the S3 subset with subsequent accumulation of tetraploid cells. Interestingly, physiological endoreduplication is initiated by p21 and E2Fs transcription factors, and Mcph1 functionally interacts with E2f1. Our findings suggest that, in the absence of Mcph1, Cdkn1a overexpression possibly combined to a decreased E2f1 activity may lead to endoreduplication in S3 progenitors, impairing further differentiation into mature red blood cells. Few data are available for patients with MCPH1 mutations, hematological defects being possibly outlooked due to the severity of the neurological phenotype. However, CBC performed in one of our patients revealed a macrocytosis consistent with dyserythropoiesis evidenced in mice. Conclusion: We demonstrate for the first time that Mcph1 expression is critical during terminal erythroid differentiation in mice. Altogether our findings provide additional evidence of a unique link between hematopoiesis and neuronal development. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 935-935
Author(s):  
Jinchang Zhao ◽  
Shinji Kondo ◽  
Sofya Suidasari ◽  
Miki Yokozawa ◽  
Ken Yamauchi ◽  
...  

Abstract Introduction Anemia is the most common blood disorder which affects billions of people, especially young adults and women. However, currently available treatments of anemia are with limitations and adverse effects. Therefore, natural resources have been receiving considerable attention as complementary or alternative hematinic agents in recent years. In this regard, olive leaf extract (OLE) is rich in bioactive phenolic compounds and has been reported to have anti-inflammatory, antioxidant and neuroprotective effects (Vogel et al., 2015). Our previous study showed treatment with water extract of olive leaf (WOL) for 12 days could induce erythroid differentiation in human hematopoietic stem cells (hHSCs) and showed potential in oxygen and iron homeostasis, haem metabolism, and hemoglobin (Hb) biosynthesis (Kondo et al., 2021). In the present study, we aimed to investigate the therapeutic effects of WOL on the phenylhydrazine (PHZ)-induced mice model of anemia and explore its underlying molecular mechanism. Methods Total 63 male ICR mice (four weeks old) were randomly divided into three groups: control, PHZ and WOL groups. Mice in the WOL group were orally administered with 150 mg/kg body weight of WOL (diluted by saline) every day, while mice in the other two groups received saline. After two weeks of WOL pretreatment, PHZ was injected intraperitoneally (60 mg/kg body weight) to PHZ and WOL groups. The mice were dissected on day0 (before PHZ injection), day1, day3, day5, and day7 after PHZ injection. We collected blood for hematologic tests during dissection and liver, kidney, intestine, bone marrow, and spleen samples. We next checked the iron homeostasis-related gene expressions by real-time PCR: hepcidin (Hamp) in liver, ferroportin (Fpn) in spleen and intestine. Results and Discussion After anemia was induced, WOL group showed a significant increase in the number of reticulocytes, the erythroid progenitors, compared to that of in PHZ group on Day 5. Simultaneously, plasma iron level on Day 5 and Hb level on Day7 was significantly decreased in the WOL group compared to the PHZ group. These results suggest that during anemia, the WOL group had rapid erythropoiesis, which in turn caused a rapid consumption of Hb and iron. Additionally, mRNA expression of Hamp in liver was significantly decreased on Day 5, whereas Fpn expressions in spleen and intestine were significantly increased on Day 5 and Day 7 in the WOL group compared to the PHZ group. Hamp regulates plasma iron concentrations as well as systemic iron metabolism by interacting with its receptor Fpn, a transmembrane iron-exporter. In intestine, Fpn controls iron absorption from food intake, while in spleen, Fpn regulates phagocytosis of senescent erythrocytes by the macrophages. Decreased Hamp expression and increased Fpn expression by WOL treatment suggest increased levels of absorbed and recycled iron to meet the demand for erythropoiesis. Altogether, our findings indicate that WOL could regulate iron homeostasis in the early stage of anemia to promote the differentiation of erythroid progenitors. Conclusion Our study suggests that WOL treatment downregulates Hamp expression in liver and increased Fpn expressions in the intestine and spleen, leading to increased absorbed iron in the intestine and recycled iron in the spleen (Figure 1). Therefore, WOL may have therapeutic potential in anemia through regulating iron homeostasis and promoting erythroid differentiation. However, further studies are required to confirm these findings and to elucidate the molecular mechanisms. Our previous study showed that 12-day treatment of hHSCs with WOL could upregulate the expression of hypoxia-inducible factor 1-a (HIF1A) (Kondo et al., 2021), a key modulator of the transcriptional response to hypoxia and has been reported to regulate erythropoietin (Epo) production, which plays an important role in erythropoiesis. So, we hypothesize that WOL treatment would activate HIF1A in anemia-induced mice model, thus regulating Epo production to accelerate erythroid maturation. HIF2A is also reported to regulate Epo in kidney and Fpn in intestine to regulate erythropoiesis (Andrew J., et al., 2019). Therefore, we will investigate Epo expression in kidney, Hif1a and Hif2a expressions in kidney and intestine, and transferrin receptor (Tfrc, erythroid marker) in bone marrow. All gene expressions will be confirmed in protein levels also. Figure 1 Figure 1. Disclosures Suidasari: Nutrition Act Co. Ltd.: Current Employment. Yokozawa: Nutrition Act Co. Ltd.: Current Employment. Yamauchi: Nutrition Act Co. Ltd.: Current Employment.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 920-920
Author(s):  
Elena C. Brindley ◽  
Emily Hartman ◽  
Julien Papoin ◽  
Brian Dulmovits ◽  
Steven L. Ciciotte ◽  
...  

Abstract RASA3, a Ras GTPase activating protein, is critical to vertebrate erythropoiesis and megakaryopoiesis. The autosomal recessive mouse model scat (severe combined anemia and thrombocytopenia) carries a G125V mutation in Rasa3 that leads to profound bone marrow failure with characteristics of aplastic anemia. The phenotype is cyclic, and mice alternate between periods of crisis and remission. Our previous studies demonstrated that this mutation in Rasa3 causes defects in several aspects of erythropoiesis, including a significant delay of erythroid differentiation at the polychromatophilic stage, decreased hemoglobinization, defects in cell cycle progression past the G1 checkpoint, and increased reactive oxygen species (ROS) during terminal erythroid differentiation as well as in scat peripheral blood reticulocytes and red blood cells. We previously reported that the mislocalization of mutated RASA3 to the cytosol of reticulocytes and mature red cells plays a role in the erythropoietic defect in scat, and the observed cell cycle arrest and increased ROS likely also contribute to this unique disease phenotype. Our current efforts are focused on further elucidation of the mechanism and specific disruptions in Ras signaling that lead to anemia, membrane fragmentation, and the cyclic phenotype in scat. Interestingly, we report here that apoptosis is not increased during scat crisis, and that mitochondria, a potential source of ROS, are normally eliminated at the reticulocyte stage. The dramatic nature of remission, with complete normalization of all hematologic parameters, led us to hypothesize that a secreted factor may be mediating the cyclic phenotype of scat. Differences in the cytokine profile of the serum of scat mice compared to wild type suggest that, indeed, one or several secreted factor(s) may be influencing the occurrence of bone marrow failure. Levels of galectin-1, a known mediator of cell-cell interactions and intracellular signaling in the hematopoietic niche, are consistently decreased in scat serum according to a multispot anti-cytokine antibody array (23,326.5 ± 21,439.7 integrated density in scat vs. 31,019.6± 20,110.7 in controls, p&lt;0.05).Studies exploring the influence of the galectin family on erythropoiesis and Ras signaling in the context of scat are underway. Strengthening the notion that RASA3 has a critical conserved role in vertebrate terminal erythropoiesis, the characteristics of bone marrow failure seen in scat have been reproduced in human CD34+ cells using siRNA and shRNA knockdowns of Rasa3 . Similar to the changes seen in scat, cells with decreased RASA3 demonstrated delayed terminal erythroid differentiation and defective hemoglobinization. Finally, analysis of Ras expression and functional pull-down studies in human CD34+ cells revealed that, while K-Ras is the major active isoform expressed during terminal erythroid differentiation, H-Ras is also active during human erythropoiesis. Future studies with CD34+ Rasa3 knockdown cells will explore the influence of RASA3 on human K- and H-Ras signaling in erythropoiesis. Taken together, our studies further characterize the vital role of RASA3 in hemoglobinization, cell cycle progression, and cell survival during terminal erythroid differentiation, as well as identify novel targets for investigation of unknown mechanisms (e.g., dysregulated cytokine secretion) of bone marrow failure syndromes. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Joschka Heil ◽  
Victor Olsavszky ◽  
Katrin Busch ◽  
Kay Klapproth ◽  
Carolina de la Torre ◽  
...  

AbstractWithin the bone marrow microenvironment, endothelial cells (EC) exert important functions. Arterial EC support hematopoiesis while H-type capillaries induce bone formation. Here, we show that BM sinusoidal EC (BM-SEC) actively control erythropoiesis. Mice with stabilized β-catenin in BM-SEC (Ctnnb1OE-SEC) generated by using a BM-SEC-restricted Cre mouse line (Stab2-iCreF3) develop fatal anemia. While activation of Wnt-signaling in BM-SEC causes an increase in erythroblast subsets (PII–PIV), mature erythroid cells (PV) are reduced indicating impairment of terminal erythroid differentiation/reticulocyte maturation. Transplantation of Ctnnb1OE-SEC hematopoietic stem cells into wildtype recipients confirms lethal anemia to be caused by cell-extrinsic, endothelial-mediated effects. Ctnnb1OE-SEC BM-SEC reveal aberrant sinusoidal differentiation with altered EC gene expression and perisinusoidal ECM deposition and angiocrine dysregulation with de novo endothelial expression of FGF23 and DKK2, elevated in anemia and involved in vascular stabilization, respectively. Our study demonstrates that BM-SEC play an important role in the bone marrow microenvironment in health and disease.


Sign in / Sign up

Export Citation Format

Share Document