scholarly journals Erythroid precursors and progenitors suppress adaptive immunity and get invaded by SARS-CoV-2

Author(s):  
Shima Shahbaz ◽  
Lai Xu ◽  
Mohammad Osman ◽  
Wendy Sligl ◽  
Justin Shields ◽  
...  

AbstractSARS-CoV-2 infection is associated with lower blood oxygen levels even in patients without hypoxia requiring hospitalization. This discordance illustrates the need for a more unifying explanation as to whether SARS-CoV-2 directly or indirectly affects erythropoiesis. Here we show significantly enriched CD71+ erythroid precursors/progenitors in the blood circulation of COVID-19 patients that have distinctive immunosuppressive properties. A subpopulation of abundant erythroid cells, CD45+CD71+cells, co-express ACE2, TMPRSS2, CD147, CD26 and these can be infected with SARS-CoV-2. In turn, pre-treatment of erythroid cells with dexamethasone significantly diminished ACE2/TMPRSS2 expression and subsequently reduced their infectivity with SARS-CoV-2. Taken together, pathological abundance of erythroid cells might reflect stress erythropoiesis due to the invasion of erythroid progenitors by SARS-CoV-2. This may provide a novel insight into the impact of SARS-CoV-2 on erythropoiesis and hypoxia seen in COVID-19 patients.

Blood ◽  
2009 ◽  
Vol 113 (16) ◽  
pp. 3845-3856 ◽  
Author(s):  
Amy L. Samuels ◽  
S. Peter Klinken ◽  
Evan Ingley

Abstract Erythropoiesis is primarily controlled by erythropoietin (Epo), which stimulates proliferation, differentiation, and survival of erythroid precursors. We have previously shown that the tyrosine kinase Lyn is critical for transducing differentiation signals emanating from the activated Epo receptor. A yeast 2-hybrid screen for downstream effectors of Lyn identified a novel protein, Liar (Lyn-interacting ankyrin repeat), which forms a multiprotein complex with Lyn and HS1 in erythroid cells. Interestingly, 3 of the ankyrin repeats of Liar define a novel SH3 binding region for Lyn and HS1. Liar also contains functional nuclear localization and nuclear export sequences and shuttles rapidly between the nucleus and cytoplasm. Ectopic expression of Liar inhibited the differentiation of normal erythroid progenitors, as well as immortalized erythroid cells. Significantly, Liar affected Epo-activated signaling molecules including Erk2, STAT5, Akt, and Lyn. These results show that Liar is a novel Lyn-interacting molecule that plays an important role in regulating intracellular signaling events associated with erythroid terminal differentiation.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2196-2196
Author(s):  
Roberta Chessa ◽  
Ritama Gupta ◽  
Bart J Crielaard ◽  
Carla Casu ◽  
Rick Feldman ◽  
...  

Abstract After blood loss, the production of red cells must be increased by stress erythropoiesis. This phenomenon is associated with increased proliferation and reduced differentiation of the erythroblasts, leading to a net increase in the number of progenitor erythroid cells and red cells (erythron). In normal conditions, after expansion of the pool of erythroblasts, these cells eventually differentiate to erythrocytes and the anemia resolves. However, in diseases such as β−thalassemia, production of healthy mature erythrocytes is impaired, resulting in anemia. Over time, the expansion, rather than the differentiation, of the erythron further exacerbates the ineffective erythropoiesis (IE), reducing the ability of the erythroid progenitors to generate erythrocytes. Interrupting the interaction between macrophages and erythroblasts (MEI) in thalassemia models is efficacious in reducing IE and alleviating the disease phenotype. We speculate that these molecules are also responsible for the homing of erythroid progenitor cells to extramedullary organs, such as the spleen and liver. Our studies in erythroblasts indicate that integrin beta−1 (Itgβ1) and also intracellular molecules such as focal adhesion kinase (Fak1), Talin−1 and Sharpin might play a role in stress erythropoiesis. Furthermore, there is increased interaction between Itgb1 and Fak1 in erythroblasts co−cultured with macrophages as demonstrated by immunocytochemistry and in vitro proximity ligation assays. In addition, targeting either Itgβ1 or Fak1 prevents expansion of erythroid cells when cultured in the presence of macrophages. Strikingly, using Itgβ1 together with Ter119 as selection parameters in flow cytometry, a distinct subset of erythroblasts, not discernable using CD44 or CD71, was observable, which we found to be part of the mixed orthochromatic erythroblast/reticulocyte population as determined with CD44 expression. Enucleation of erythroblasts was accompanied by a marked loss of Itgβ1 expression, indicating that Itgβ1 may be involved in erythroblast enucleation and differentiation. We crossed Hbbth3/+ mice with animals in which Itgβ1 or Fak1 were floxed and carrying an inducible Cre−recombinase (Mx1−Cre). From these animals, we investigated three different models; two obtained from breeding (Hbbth3/+−Itgβ1fl/fl−Mx1−Cre and Hbbth3/+−Fak1fl/fl−Mx1−Cre) and one by bone marrow transplant (BMT) of hematopoietic stem cells (HSCs) of Hbbth3/+−Itgβ1fl/fl −Mx1−Cre animals into wt mice to generate thalassemic animals that expressed the floxed Itgβ1 only in hematopoietic cells. After serial administration of Poly(I)−Poly(C) [poly(I:C)] the animals were analyzed for their erythropoiesis in the bone marrow and spleen. All the animals treated with poly(I:C) showed populations of Itgβ1 or Fak1 negative cells in the bone marrow and spleen. This indicated that all the HSCs were successfully depleted of the Itgβ1 or Fak1 gene. Interestingly, the spleen weight of all the treated animals was reduced, on average, 50% compared to untreated thalassemic mice. Similar results were seen also in Hbbth3/+−Itgβ1fl/fl−Mx1−Cre animals generated through BMT. Therefore, Itgβ1 and Fak1 might contribute to the pathophysiology of thalassemia and their removal might result in reduced stress erythropoiesis, erythroid proliferation and, as a consequence, amelioration of splenomegaly. Iron analysis and quantification of Erythroferrone (ERFE) are in progress to evaluate the impact of depleting Itgβ1 and Fak1 on these mechanisms. We are now in the process of identifying compounds that target MEI and, in particular, Itgβ1. Such molecules might be utilized for development of new treatments for thalassemia or additional disorders of aberrant erythropoiesis. Disclosures Feldman: Bayer ealthCare Phamaceuticals Inc.: Employment. Rivella:isis Pharmaceuticals: Consultancy; Merganser Biotech: Other: Stock options; Novartis Pharmaceuticals: Consultancy; Medgenics Pharmaceuticals: Consultancy; Bayer Healthcare: Consultancy, Research Funding.


2020 ◽  
Author(s):  
Shanquan Wang ◽  
Zhiwei Liang ◽  
Jiangjian Shi ◽  
Chen Wang ◽  
Junhui Li ◽  
...  

Abstract Background: Wastewater treatment generates large amounts of waste activated sludge (WAS), which mainly consist of recalcitrant microbial cells and particulate organic matter. WAS pre-treatment is an effective way to destabilize sludge floc structure and release cellular macromolecules and other organic matter for improvement of digestion efficiency. Nonetheless, impacts of WAS pre-treatment on the complex digestion sludge microbiome, as well as mechanistic insight into how sludge pre-treatment improve digestion performance, remain to be elucidated. Results: In this study, genome-centric metagenomic approach was employed to investigate the digestion sludge microbiome in four methanogenic sludge digesters with different feeding sludge: APAD, WAS pre-treated with 0.25 mol/L alkaline/acid; HS-APAD, WAS pre-treated with 0.8 mol/L alkaline/acid; Thermal-AD, thermal pre-treated WAS; Control-AD, fresh WAS. We retrieved 254 metagenomic-assembled genomes (MAGs) to identify the key functional populations involved in methanogenic digestion process. These MAGs span 28 phyla with 69 of them as yet-to-be-cultivated lineages, and 30 novel lineages were characterized with metabolic potential associated with hydrolysis and fermentation. Interestingly, functional populations involving carbohydrate digestion were overrepresented in APAD and HS-APAD, while lineages related to protein and lipid fermentation were overrepresented in Thermal-AD, reflecting different digestion substrates released from alkaline/acid and thermal pre-treatments. Of the three major functional populations, i.e., fermentative bacteria, acetogenic syntrophs and methanogens, significant correlations between genome sizes of the fermentative bacteria and their abundance were observed, particularly in the APAD and HS-APAD with improved digestion performance. Conclusion: These genome-centric metagenomic insights advance our understanding of sludge pre-treatment on digestion sludge microbiomes, shedding light on future optimization of methanogenic sludge digestion and resource recovery.


Blood ◽  
2008 ◽  
Vol 112 (3) ◽  
pp. 470-478 ◽  
Author(s):  
Joel Anne Chasis ◽  
Narla Mohandas

Abstract Erythroblastic islands, the specialized niches in which erythroid precursors proliferate, differentiate, and enucleate, were first described 50 years ago by analysis of transmission electron micrographs of bone marrow. These hematopoietic subcompartments are composed of erythroblasts surrounding a central macrophage. A hiatus of several decades followed, during which the importance of erythroblastic islands remained unrecognized as erythroid progenitors were shown to possess an autonomous differentiation program with a capacity to complete terminal differentiation in vitro in the presence of erythropoietin but without macrophages. However, as the extent of proliferation, differentiation, and enucleation efficiency documented in vivo could not be recapitulated in vitro, a resurgence of interest in erythroid niches has emerged. We now have an increased molecular understanding of processes operating within erythroid niches, including cell-cell and cell-extracellular matrix adhesion, positive and negative regulatory feedback, and central macrophage function. These features of erythroblast islands represent important contributors to normal erythroid development, as well as altered erythropoiesis found in such diverse diseases as anemia of inflammation and chronic disease, myelodysplasia, thalassemia, and malarial anemia. Coupling of historical, current, and future insights will be essential to understand the tightly regulated production of red cells both in steady state and stress erythropoiesis.


2019 ◽  
Vol 3 (14) ◽  
pp. 2205-2217 ◽  
Author(s):  
Siyang Hao ◽  
Jie Xiang ◽  
Dai-Chen Wu ◽  
James W. Fraser ◽  
Baiye Ruan ◽  
...  

Abstract Anemic stress induces the proliferation of stress erythroid progenitors in the murine spleen that subsequently differentiate to generate erythrocytes to maintain homeostasis. This process relies on the interaction between stress erythroid progenitors and the signals generated in the splenic erythroid niche. In this study, we demonstrate that although growth-differentiation factor 15 (Gdf15) is not required for steady-state erythropoiesis, it plays an essential role in stress erythropoiesis. Gdf15 acts at 2 levels. In the splenic niche, Gdf15−/− mice exhibit defects in the monocyte-derived expansion of the splenic niche, resulting in impaired proliferation of stress erythroid progenitors and production of stress burst forming unit-erythroid cells. Furthermore, Gdf15 signaling maintains the hypoxia-dependent expression of the niche signal, Bmp4, whereas in stress erythroid progenitors, Gdf15 signaling regulates the expression of metabolic enzymes, which contribute to the rapid proliferation of stress erythroid progenitors. Thus, Gdf15 functions as a comprehensive regulator that coordinates the stress erythroid microenvironment with the metabolic status of progenitors to promote stress erythropoiesis.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1035-1035 ◽  
Author(s):  
Carla Casu ◽  
Pedro Ramos ◽  
Ella Guy ◽  
Nico van Rooijen ◽  
Robert W Grady ◽  
...  

Abstract Abstract 1035 β-Thalassemia is a disorder associated with abnormal β-globin production, leading to anemia, extramedullary hematopoiesis (EMH), a decreased lifespan of the red cells and iron overload. In this disorder erythropoiesis is ineffective due to increased erythroid apoptosis and erythroblast proliferation, as well as deficient differentiation. Recent evidence suggests that erythroid development, especially under conditions of anemia (stress erythropoiesis), is highly dependent on microenvironmental factors within the erythroid niche, potentially mediated by the interaction of erythroblasts with macrophages. However, little is known about the function of these cells in pathological anemias associated with abnormal erythropoiesis. Our goal was to study the role of macrophages in normal, stress and ineffective erythropoiesis (IE). Macrophages were eliminated by intravenous administration of clodronate-containing liposomes. Treatment was carried out for up to 12 weeks, serial measurements being made of erythropoietic and pathological parameters. As a model of stress erythropoiesis, phlebotomized wt mice were used. To study IE we utilized th3/+ mice, a model of β-thalassemia intermedia (TI). Clodronate treatment effectively depleted splenic and bone marrow (BM) macrophages as shown by FACS and immunohistochemical analyses. Depletion of macrophages in wt mice had little effect on steady state erythropoiesis. In contrast, clodronate treatment drastically impaired the response to stress erythropoiesis in these mice, as shown by the slow recovery from phlebotomy-induced anemia. This was associated with a very slow rate of RBC and reticulocyte production, suggesting that erythroid activity was markedly impaired. Accordingly, mice depleted of macrophages were unable to expand their pool of erythroid progenitors in the BM and spleen in response to anemia, suggesting that macrophages play a critical role in this process. A similar defect was observed in response to Epo stimulation, suggesting that an intact erythroid niche is essential for normal activity of Epo in promoting erythroid expansion. Interestingly, TI mice treated with clodronate exhibited an improvement of the thalassemic phenotype. Within 40 hours of clodronate treatment, mice showed an increase in hemoglobin (Hb), RBC and reticulocyte counts in the peripheral blood, and a reduction of extra-medullary hematopoiesis (increased ratio of mature to immature erythroid cells) and splenomegaly (P<0.05 for all parameters analyzed). This indicated a more effective erythropoiesis in the absence of macrophages, suggesting that these cells negatively influence erythroid development in this disorder. Improvement of anemia was maintained for up to 12 weeks of continuous treatment, and was associated with increased RBC counts. Under these conditions, serum iron was markedly decreased, potentially reducing iron delivery to maturing RBCs. Recent studies have suggested that lowering iron delivery to erythroblasts leads to improvement of the RBC phenotype in this disorder. Consistently, MCHs were decreased after macrophage depletion, which correlated with lower accumulation of alpha-globin/heme precipitates in the RBC membranes. Moreover, the RBC lifespan in clodronate-treated mice was increased compared to that in PBS controls. This difference was maintained even when these cells were transfused into wt mice, suggesting that it was not associated exclusively with deficient RBC clearance in macrophage-depleted mice. In conclusion, our data suggests that macrophages have two major roles in β-thalassemia: 1) to modulate iron availability for erythroid cells; 2) to impair erythroid development, as suggested by the amelioration of splenomegaly and EMH observed after clodronate treatment. We hypothesize that the macrophages within erythroblastic islands control erythropoiesis, acting as modulators of this process. Under conditions of stress erythropoiesis they positively influence erythroid development, promoting proliferation to increase the pool of erythroid cells. However, under conditions of chronic stress such as in TI, macrophages limit differentiation and promote excessive expansion of the erythron, contributing to IE. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 31-32
Author(s):  
Parul Rai ◽  
Swarnava Roy ◽  
Diamantis G. Konstantinidis ◽  
Sithara Raju Ponny ◽  
Marthe-sandrine Eiymo Mwa Mpollo ◽  
...  

Sickle cell anemia (SCA), caused by a mutant β-globin gene, results in polymerization of the abnormal hemoglobin S and sickle shaped RBCs that cause vascular occlusions, chronic hemolytic anemia and cumulative organ damage. We have reported sickle nephropathy from elevated levels of angiotensin-II (AT), the bioactive peptide of the renin-angiotensin system (RAS), by signaling through the AT receptor-1 (AT1R), in both mice and humans with SCA, and that occurs in the absence of hypertension or hyper-reninemia [Roy et al, AJH, 2018]. Herein, we investigated the mechanisms underlying the hyperangiotensinemia in SCA. We found that AT levels are elevated due to higher oxidized state of its precursor, angiotensinogen (ATGN), which results from the elevated reactive oxygen species (ROS) in SCA. Oxidized ATGN is more rapidly converted to AT. Hence, the high ROS in SCA increases AT production. Blockade of AT-AT1R signaling in SCA mice, either globally (with captopril that reduces AT production, or losartan that blocks AT1R, or by constitutive genetic knockout of AT1R in SCA mice [SCA AT1R-/-]), or in erythroid cells by an erythroid-specific AT1R knockout (AT1Rf/f EpoR Cre+ termed eCre+) in SCA mice (SCA eCre+) resulted in significant reduction in RBC ROS (Figure 1a). These data show that AT-AT1R signaling in turn generates ROS in sickle erythroid cells, thus creating a positive feedback loop of ↑ROS --&gt;↑RAS --&gt;↑ROS, which causes, and perpetuates the hyperangiotensinemia seen in SCA. The AT-mediated ROS-RAS loop is driven largely by sickle erythropoiesis: SCA eCre+ mice have reduced RBC ROS, hence have reduced AT, and do not develop nephropathy. Surprisingly, while global AT1R deficiency in WT mice (WT AT1R-/-) had no RBC phenotype, SCA AT1R-/- mice developed profound anemia, suggesting AT signaling may be important for the stress erythropoiesis (Stress-E) state, present in SCA. AT is known for its role as a renal erythropoietin (Epo) secretagogue, but its role in Stress-E is unknown. Induction of Stress-E in WT AT1R-/- mice (with phenylhydrazine or daily phlebotomies) resulted in higher anemia in WT AT1R-/- mice than in WT AT1R+/+ mice, suggesting AT1R signaling is important for Stress-E, not baseline erythropoiesis (Base-E). However, WT mice with erythroid-specific deficiency of AT signaling (WT eCre+), when stressed, were able to maintain hemoglobin comparable to their controls (WT eCre-), with an exponential increase in Epo level. Epo levels were high in SCA mice to begin with but were insufficient to compensate with loss of AT signaling in SCA AT1R-/- mice, resulting in development of anemia. RNAseq analysis of sorted Stress-E nucleated erythroid precursors and enucleated erythrocytes in WT AT1R-/- mice showed a remarkable downregulation of the Hedgehog, BMP4, KIT and WNT Stress-E signaling pathways, when compared to WT (AT1R+/+) mice, further confirming that AT signaling is critical, and conceivably upstream to these established Stress-E pathways. However, AT-AT1R signaling in Stress-E states is a double-edged sword: on one hand it was essential to sustain erythropoiesis, while on the other hand it resulted in significant ROS production. This was seen in SCA, and in WT mice where Stress-E was associated with higher RBC ROS (Figure 1a). AT is known to activate NADPH oxidase (NOX), to generate ROS in other cell types. But pharmacological inhibition of NOX signaling, or genetic deficiency of Gp22phox (a common subunit of NOX isoforms) in SCA mice did not lower RBC ROS. However, AT-AT1R signaling in Stress-E increased the mitochondrial mass in erythroid precursors, and also resulted in retention of dysfunctional mitochondria with lower membrane potential in the enucleated erythrocytes, a source of high ROS (Figure 1b-c). Hence, AT signaling in Stress-E inhibited mitophagy, which is required for clearance of mitochondria after enucleation. The transcriptome profile corroborated these findings: With Stress-E, WT AT1R-/- mice had upregulation of genes involved in mitophagy, and genes involved in maintaining mitochondrial integrity/quality and cellular redox homeostasis under oxidative stress. Taken together, our results show that AT signaling plays a critical role in increasing erythroid cell mass during Stress-E, but also downregulates mitophagy, antioxidant genes, and results in increased retention of dysfunctional mitochondria, which are the source of high RBC ROS that mediates end-organ injury. Disclosures Kalfa: Forma Therapeutics, Inc: Research Funding; Agios Pharmaceuticals, Inc: Consultancy, Research Funding. Malik:Aruvant Sciences, Forma Therapeutics, Inc.: Consultancy; Aruvant Sciences, CSL Behring: Patents & Royalties.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3190-3190 ◽  
Author(s):  
Monika Priwitzerova ◽  
Dagmar Pospisilova ◽  
Karel Indrak ◽  
Prem Ponka ◽  
Vladimir Divoky

Abstract Disorders of iron metabolism and homeostatis belong to the most common diseases in humans; however, neither the regulation of iron metabolism in erythroid cells nor the pathophysiology of these diseases are completely understood. Last year we reported at this meeting a case of severe hypochromic microcytic anemia caused by a homozygous mutation in the divalent metal transporter 1 (DMT1) gene (DMT1 1285G>C). The defective growth of this patient’s erythroid progenitors in vitro was rescued by the addition of 10 μM iron-salicylaldehyde isonicotinoyl hydrazone (Fe-SIH), an iron chelate that has been shown to deliver iron for heme synthesis without involving the transferrin receptor/DMT1 pathway. To further characterize the impact of the DMT1 mutation on the in vitro growth of erythroid progenitors, 10 μM Fe-SIH, 25 μM hemin or 10 μM zinc chloride (ZnCl2) were added to the erythropoietin (Epo) containing cultures. All substances increased the plating efficiency in normal control as well as mutant BFU-Es. Moreover, all substances led to a 1.4 to 1.6 fold increase in hemoglobin (Hb) content in normal BFU-Es. Although hemin did not significantly increase Hb content in the patient’s BFU-E, the addition of Fe-SIH to the Epo/hemin-containing cultures increased Hb content 2.2 fold. Since Hb synthesis in heme-deficient erythroid cells is inhibited by heme-regulated inhibitor (HRI) kinase, which specifically phosphorylates the α subunit of the eukaryotic initiation factor 2 (eIF2α), we have analyzed the level of eIF2α phosphorylation in cytospine-harvested BFU-Es. Immunofluorescence analysis of erythroid cells with phospo-eIF2α (Ser51) antibody revealed increased phosphorylation of eIF2α in mutant BFU-Es compared to control BFU-Es grown under all aforementioned conditions. The most striking differences were observed in Epo and Epo/hemin containing cultures, in which the fractions of phospho-eIF2α-positive cells were significantly higher in mutant BFU-Es. These data suggest that the block of terminal differentiation/hemoglobinization in the DMT1-mutant erythroid cells is only partially caused by translational arrest of globin chain synthesis; the delivery of iron may be required for other cellular functions apart from Hb synthesis. Interestingly, the most prominent positive effect on the proliferation (i.e., BFU-E cellularity) as well as hemoglobinization of mutant BFU-Es had a combination of Epo/Fe-SIH and ZnCl2; this combination led to the highest increase in Hb content that was associated with almost complete lack of eIF2α phosphorylation. This suggests that the addition of Zn2+ to Fe-SIH containing cultures further stimulates processes that are initially rescued in DMT1-mutant erythroid cells by the addition of iron. We conclude, that the DMT1 defect in erythroid cells is complex, probably involving the combination of transcriptional and translational defects that cannot be rescued by hemin alone. Figure Figure


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3870-3870
Author(s):  
Sathish Kumar Mungamuri ◽  
Saghi Ghaffari

Abstract Erythropoietin (Epo) signaling is required for differentiation of erythroid progenitors to mature red blood cells. Binding of Epo to its receptor activates Jak2, which in turn activates many signaling proteins including AKT, MAPK proteins and STATs. We have shown previously that AKT is required for Epo regulation of erythroid cell maturation; activated AKT complements Epo receptor signaling in JAK2-deficient fetal liver cells and supports erythroid cell differentiation. AKT functions by phosphorylating several proteins including FoxO3 and mTOR. AKT phosphorylation of FoxO3 represses FoxO3’s activity, whereas AKT-dependant phosphorylation activates mTOR and its downstream target p70 S6 kinase (S6K). We have shown recently that FoxO3 is essential for the regulation of erythroid cell cycling, maturation, lifespan and anti-oxidant response (Marinkovic et al., JCI, 2007). Here we aimed at identifying other proteins in AKT signaling network that may regulate the maturation of erythroid progenitors. To address this, we inhibited several signaling pathways and analyzed their role in Epo-dependant maturation of freshly-isolated E14 fetal liver progenitors. As anticipated, blocking PI3-Kinase resulted in 60 % reduction of BFU-E- and CFU-E-derived colony formation and blocked the maturation of erythroid progenitors. Interestingly, blocking either p38 or ERK MAPK signaling showed 40% reduction in erythroid BFU-E- and CFU-E-derived colony formation. Surprisingly, blocking of mTOR signaling inhibited the formation of BFU-E- and CFU-E-derived colonies by 75 %. Further analysis by flow cytometry monitoring of cell surface markers CD71 and TER 119 showed that erythroid progenitor cell maturation could not proceed past early erythroblast stage when cells were cultured in the presence of rapamycin overnight. We confirmed that this block in differentiation was not due to apoptosis of erythroid cells. Since both FoxO3 and mTOR work downstream of AKT, we asked whether inhibition of mTOR has any impact on FoxO3 activity. Epo stimulation of freshly isolated bone marrow lineage-negative cells previously starved from cytokines showed a 2.3 fold increase in FoxO3 phosphorylation in the presence of rapamycin, suggesting cross talk between mTOR and FoxO3. Next, we investigated the effect of loss of FoxO3 on AKT/mTOR signaling in erythroid precursors. To address this, we prepared a population of bone marrow depleted from lineage-restricted cells and cultured under optimum erythroid conditions that generated 60% erythroblasts after 18 hours. Epo stimulation of FoxO3 null erythroid precursors led to hyperphosphorylation of Jak2, AKT, mTOR and S6K as compared to control cells. Since FoxO3 is critical for repression of reactive oxygen species (ROS), we evaluated the potential role of ROS in activating these proteins in FoxO3 mutant erythroid cells. In vitro treatment with ROS scavenger N-Acetyl-Cysteine (NAC) reduced significantly the hyper-phosphorylation of AKT, mTOR and S6K in FoxO3 null erythroid precursors in response to Epo. In addition, our results suggest that phosphorylation of JAK2 and its downstream signaling proteins AKT/mTOR/S6K in primary wild type erythroid precursor cells in response to Epo is mediated by ROS. Interestingly, ROS modulation of phosphorylation of mTOR/S6K was significantly stronger than that of AKT in response to Epo-stimulation of primary erythroid cells. Activation of AKT/mTOR/S6K is likely to mediate increased production of erythroid precursors observed in FoxO3 mutant mice (Marinkovic et al., JCI, 2007). Collectively these results indicate an important function for the AKT/mTOR/S6K signaling pathway in Epo-dependant erythropoiesis and suggest that cytokine-mediated production of ROS plays a critical role in the regulation of primary erythroid cell formation.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 943-943 ◽  
Author(s):  
Genís Campreciós ◽  
Jeffrey Barminko ◽  
Jeffrey Bernitz ◽  
Saghi Ghaffari

Abstract Erythropoiesis in adult mice occurs principally in the bone marrow, while stress erythropoiesis is mostly localized in the spleen. Although both bone marrow and spleen produce fully functional mature red blood cells, we noticed that the rate of maturation of erythroid precursors (based on the ratio of mature/immature erythroid cells) is over 10 times higher in the spleen as compared to the bone marrow in C57BL6 mice (3.7 ± 0.6 in the spleen as compared to 0.4 ± 0.1 in the bone marrow, n ≥ 10 mice). Cell cycle analysis of erythroid precursors revealed that bone marrow erythroid cells cycle up to 3 times more than their spleen counterparts (88 ± 1% vs 25 ± 7% of cells in the S phase, n=3). As reactive oxygen species (ROS) influence cell cycle, we measured ROS levels by flow cytometry using the CM-H2DCFDA probe. To our surprise, we found ROS levels to decrease (rather than increase) progressively in the bone marrow erythroid cells as they mature and accumulate hemoglobin. Interestingly, the levels of ROS were twice as high in the spleen erythroid cells as compared to erythroid cells in the bone marrow. As mitochondria are a major site of ROS production we measured mitochondrial mass by flow cytometry using Mitotracker Green. Mitochondrial mass was found to be two fold lower in the spleen erythroid cells as compared to the bone marrow. In agreement with these findings, qRT-PCR expression analysis of different antioxidant enzymes such as gluthathione peroxidases Gpx1 and Gpx4 showed higher levels in the bone marrow as compared to the spleen erythroid precursors. In particular, Gpx1 expression increased ten fold during erythroid maturation in the bone marrow while in the spleen the expression of Gpx1 did not change significantly. Together these results suggest erythroid metabolic profile is distinct in the spleen as compared to the bone marrow at the steady state. In order to compare homeostatic versus stress erythropoiesis we analyzed bone marrow and spleen from Foxo3-/- and Th3/+ thalassemic mice, two models of ineffective erythropoiesis with different degrees of severity and splenomegaly. As anticipated Foxo3-/- and Th3/+ erythroid precursors displayed decreased rate of maturation as compared to wild type cells in both bone marrow (0.3 ± 0.02 and 0.12 ± 0.01 for Foxo3-/- and Th3/+ respectively, n ≥ 10) and spleen (2.1 ± 0.3 and 0.42 ± 0.1 for Foxo3-/- and Th3/+ respectively, n ≥ 10 mice per group) and cell cycle analysis showed an increased number of cells in the S phase in both Foxo3-/- and Th3/+ spleen erythroid cells as compared to wild type (63 ± 2% and 79 ± 2% respectively, n=3). Unexpectedly however, ROS levels in both Foxo3-/- and Th3/+ spleen erythroid cells were decreased as compared to their wild type counterparts. The observed inverse correlation between cell cycling and ROS levels was further supported by expression analysis of Gpx1 and Gpx4, the levels of which were increased in Foxo3-/- as compared to wild type spleen erythroid cells. Collectively our results highlight the different erythroid metabolic conditions in the bone marrow versus spleen under both homeostatic and disease states. Further investigations elucidating differences in metabolic conditions and properties of erythroid cells in bone marrow versus spleen should improve our understanding of generation of erythroid cells under stress and the production of erythroid cells in vitro. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document