Abstract
In vitro surrogate models of human erythropoiesis made many contributions to our understanding of the extrinsic and intrinsic regulation of this process in vivo and how they are altered in erythroid disorders. In the past, variability among the levels of hemoglobin F produced by adult erythroblasts generated in vitro by different laboratories identified stage of maturation, fetal bovine serum and accessory cells as “confounding factors”, i.e. parameters intrinsically wired in the experimental approach that bias the results observed. The discovery of these factors facilitated the identification of drugs that accelerate terminal maturation or activate specific signaling pathways for the treatment of hemoglobinopathies. It also inspired studies to understand how erythropoiesis is regulated by macrophages present in the erythroid islands. Recent cell culture advances have greatly increased the number of human erythroid cells that can be generated in vitro and are used as experimental models to study diseases, such as Diamond Blackfan Anemia, which were previously poorly amenable to investigation. However, in addition to the confounding factors already identified, improvement of the culture models has introduced novel confounding factors, such as possible interactions between signaling from cKIT, the receptor for stem cell factor, and from the glucocorticoid receptor, the cell proliferation potential and the clinical state of the patients. This review will illustrate these new confounding factors and discuss their clinical translation potential to improve our understanding of Diamond Blackfan Anemia and other erythroid disorders.
Keywords: Erythropoiesis, Experimental models, Anemia, Clinical Translation, Cell Culture Advances
Modelling the erythroid island in vitro
In solution, erythroblasts and macrophages tend to form associations which are resistant to modest physical stresses such as those established when the cells pass through a cytofluorimeter flow or during the centrifugation step necessary to prepare cytosmear [19,20]. The mechanisms which regulate this in vitro association are currently under investigation and may shed light on the formation of the erythroid islands in the bone marrow. This representative confocal microscopy image of a cytocentrifuged preparation depicts interactions occurring between human erythroid cells and macrophages generated in culture. The cells were obtained from a polycythemia vera patient cultured for 10 days under the same conditions used to expand those presented in the Movie and were stained with antibodies for erythroid macrophage protein (EMP) [21] (green signal) and Glycophorin A (red signal). Macrophages express robust levels of EMP while erythroblasts (identified by the Glycophorin A staining) do not. Nuclei were counterstained with DAPI. Original magnification 1500×. The representative scale bar corresponds to 10μm.
Introduction
In vivo studies on the regulation of human erythropoiesis and how it is altered in disease states is challenged by ethical considerations that limit the manipulations to which human beings may be subjected. In the 70’s, this recognition led to the development of surrogate assays for human hematopoiesis represented by semisolid cultures in which human bone marrow cells were induced to generate colonies composed of mature blood cells [1]. These assays, as imperfect as they were at that time, were instrumental to move the field forward. As examples, they provided the biological basis that guided the identification, biochemical purification and genetic cloning of human hematopoietic growth factors [2] and, coupled with genetic investigations, allowed demonstration of the clonal nature of some malignant hematopoietic disorders, such as polycythemia vera [3]. Establishing these in vitro models represented such a great success in itself that criticism on how accurately they predict in vivo situations was not fully explored. The fact that cells and proteins identified by these models exerted in vivo the expected biological functions was considered sufficient to validate their use.
First realization that in vitro models are sensitive to “Confounding Factors”
In the 80’s, the availability of erythropoietin (EPO) purified from the sera of polycythemic sheep, the by now forgotten step-III EPO [4], made possible the use of in vitro assays for identifying the abnormalities of hemoglobin production in patients with hemoglobinopathies [5]. These studies soon recognized that erythroid progenitor cells from non-diseased adult individuals have the ability at the single cell level to generate in vitro erythroid cells that produce fetal (F) hemoglobin [6]. However, the ranges of hemoglobin F detected in these cells by different laboratories varied widely (from 0–1% up to 15%) [7–9]. To reconcile this variability, a systematic analysis of the culture components was conducted out identify which of them could affect hemoglobin F production. These parameters, or as currently defined “confounding factors” [10], were represented by “stage of cell maturation”, EPO concentration and presence of macrophages and of compounds contained in selected batches of fetal bovine serum [11–14]. In particular, fetal bovine serum was recognized as a source of factors, some of which were removable by charcoal treatment, which could exert either negative or positive effects on hemoglobin F production [15–17].
The recognition that in vitro models contain intrinsic biases did not discourage some investigators who were instead inspired by them to develop novel areas of research. As examples, the confounding factor “stage of maturation” lead to design therapies for hemoglobinopathies based on drugs that accelerate terminal maturation, such as hydroxyurea [18]; the identification of the regulatory effects of macrophages fostered the development of in vitro models to study the erythroid bone marrow niche (Movie 1 and Graphical Abstract) [19–21] while the factors present in fetal bovine serum, by providing proof-of-principle that in adult cells hemoglobin switching is sensitive to micro-environmental cues, inspired studies aimed toward the identification of signal transduction elements that could be targeted to increase hemoglobin F production in patients with hemoglobinopathies [22,23].
Other investigators instead considered in vitro modeling as lacking rigor and developed holistic approaches represented by mouse models to study human hemoglobin switching that were instrumental to the identification of the regulatory regions of the human β-globin locus [24,25].
Improvement of in vitro models for human erythropoiesis
In the 90’s, the cloning of stem cell factor (SCF, also known as Steel factor or KIT ligand) [26–28], which in combination with EPO regulates red blood cell production in vivo [29], greatly facilitated our ability to model human erythropoiesis in vitro. In liquid cultures supplemented with optimal concentrations of SCF and EPO, and suboptimal concentrations of interleukin-3 (IL-3), human hematopoietic progenitor cells undergo synchronous uni-lineage differentiation which generates erythroblasts by day 10–12 and red blood cells by day 17–20 [30,31]. These cultures are considered models for steady-state hematopoiesis because growth factors are used at concentrations that, although higher than those present in human plasma, may reflect those present in the bone marrow. In the bone marrow, SCF is expressed by the niche in a membrane-bound form which is 1–2-logs more potent than the soluble form used in culture [32], IL-3 is produced locally by the adipocytes [33] and EPO establishes synergies with components of the stroma which improve its bio-availability [34]. Steady-state modeling generates 5,000–50,000 erythroid cells per progenitor cell, numbers which are sufficient for most but not all biochemical studies.
Studies in mice on the recovery from hemolytic anemia induced by phenyl-hydrazine, which has represented for years the gold standard to measure EPO concentration in biological fluids [35], led to the recognition that recovery from stress requires in addition to EPO [36] increased levels of glucocorticoids, hormones produced by the adrenal gland and released in blood [37]. Glucocorticoids exert their functions by binding to the glucocorticoid receptor (GR) and activate the stress response in all mammalian cells [37]. The clinical observation that patients with gain (Cushing’s syndrome) or loss (Addison’s disease) of GR function develop, respectively, erythrocytosis or anemia [38,39] indicated that GR regulates stress erythropoiesis also in man. This recognition inspired the development of liquid cultures stimulated with SCF, IL-3 and EPO plus a synthetic GR agonist, either hydrocortisone (HC) or dexamethasone (Dex), which is 10-fold more active, as surrogate models for human “stress erythropoiesis” [40–42]. In these models, differentiation remains uni-lineage but the erythroid cells from the BFU-E [43–45] to the pro-erythroblast stage [46], express a set of GR-induced genes (such as ZFP36L2) which retains them into a self-renewal state blocking progression to terminal maturation [43,44,47]. By day 10–14, stress-specific cultures generate erythroblasts that, although similar in morphology and pattern of CD71/CD36/CD49d/CD235a expression to those generated in cultures without glucocorticoids [48,49], represent distinctive cell populations that should be more properly defined stress-specific erythroid cells. In fact, in addition to expressing GR-induced genes [43,44,47], these cells differ from those generated in the absence of Dex for lacking MPL (the receptor for thrombopoietin) and gaining TGF-β receptor 3 expression [46,50] and for being responsive to BMP-4 [51,52] and not to thrombopoietin [46]. When transferred to differentiation cultures containing EPO, insulin, transferrin and thyroid hormone, stress-specific erythroblasts, as those obtained without Dex, mature into reticulocytes within 7 days [53]. Culture models of stress erythropoiesis generate great numbers (>105 cells per progenitor cell) of erythroid cells starting from limited volumes of blood and are therefore suited for studies involving pediatric patients with congenital erythroid disorders (see Figure 1). As an example, this model has been instrumental to define the sequela of molecular epigenetic events which ultimately leads to the production of red blood cells by sources from different developmental stages (fetal liver, cord blood and adult blood and marrow) [54,55].
Figure 1. Examples of clinical applications of in vitro modeling of erythroid cells.
Left Panel: Representative confocal microscopy image of erythroid cells expanded in vitro from adult blood stained with antibodies against calreticulin (red signal) and the glucocorticoid receptor (GR). These studies may elucidate mechanisms which regulate the stress response in human erythroid cells and how their alterations may determine the pathobiology of myeloproliferative neoplasms [90]. Right panel: Representative confocal microscopy analyses for EKLF (also known as KLF1, green signal) in erythroid cells expanded in vitro from adult blood. Human erythroid cells expanded in vitro may elucidate the cross-regulation between EKLF and BCL11A in the regulation of hemoglobin F production as well identify the effects exerted by EKLF mutations in human erythropoiesis [91]. Nuclei were counterstained with DAPI. Original magnification 1500×. Scale bars are not presented.
Rationale supporting the use of improved in vitro models to study human erythropoiesis
Restrictions posed by animal welfare activists on mouse experiments have recently revitalized the use of surrogate in vitro models to study human erythropoiesis. This use has also been fostered by the increasing numbers of reports describing significant differences in the regulation of murine and human erythropoiesis [56]. One of the first differences to be noted is the ability of granulocyte-macrophage colony stimulating factor (GM-CSF, currently known as CSF2) to stimulate erythroid differentiation in culture [57]. In human cultures, GM-SCF stimulates the growth of a subset of erythroid bursts which does not respond to IL-3 while in murine cultures it only stimulates the growth of granulo-monocytic colonies. This difference suggests that the restriction between erythroid and myeloid differentiation is more stringent in murine than in human hematopoiesis. The biochemical basis for these differences is starting to emerge. In fact, the signaling cascade of the EPO receptor includes Ca2+ mobilization and the regulator of human erythroid cell expansion (RHEX) factor in human but not in murine erythroid cells genome [58,59]. The use of improved culture models of human erythropoiesis has been favored by the development of retroviral technologies which allow altering gene expression or performing gene editing with low numbers of human stem cells.
It has been recognized that also results obtained with these more advanced in vitro models by different laboratories may be widely different. By using sophisticated bio-informatic approaches, a recent commentary by Ulirsch et al. rediscovered that “stage of maturation” is a confounding factor which may affect the interpretation of gene expression analyses and discusses how this factor may bias the interpretation of results obtained using these advanced in vitro models [10]. This commentary ends with the recommendation that conclusions must be supported by results obtained with a uniformly rigorous definition of stage of maturation. However, as pointed out by a related commentary [60], in the case of some diseases, variegation in maturation profile is to be expected because the underlying mutations may alter the expression of the antigens used for its definition. This is the case, for example, of some of the mutations in EKLF which impair expression of the Lutheran antigen [61]. Still, differences observed between these non-perfectly matched cells speak about the mechanisms which had determined the alterations observed.
In addition to maturation stage, however, advanced in vitro models contain novel “confounding factors” which are still poorly recognized and discussed which may represent both a challenge and a source of inspiration for additional studies. Below, we compared the culture technology used in the latest studies published on Diamond Blackfan Anemia (DBA) to identify some of these novel confounding factors and to discuss how they may inspire areas of research which may increase our knowledge on this, and possibly other erythroid disorders.
Novel “confounding factors” and some the avenues of research they may inspire
In muscle cells, GR activates the expression of genes encoding factors that, by suppressing mTOR, reduce phosphorylation of 4E-BP1 and S6K1, inhibiting the early steps (binding to 40S and initiation) of mRNA translation by the ribosome machinery and reducing protein synthesis [62–66]. It is therefore counterintuitive that treatment with glucocorticoids is effective in patients suffering from DBA, a congenical form of pure red blood cell aplasia induced in many cases by genetic mutations that impair the initial steps of mRNA translation by the ribosomes [67,68]. For reasons still unknown, the treatment is effective only in 60% of the patients and the identification of strategies to cure DBA represents an important unmet clinical need. To identify novel therapeutic targets, scholars have often used surrogate models of human erythropoiesis as tools to obtain a deeper understanding of the pathobiology of the disease. As examples, the discovery that SCF rescues the ability of hematopoietic progenitor cells from DBA to generate erythroid bursts in culture [69–71] inspired an international trial with recombinant SCF that was prematurely interrupted by the National Institute of Health after the death of one patient (Dr. Ramenghi, personal communication) although results obtained with some of the patients are available as case reports (see for example 72). More recently, the observation that DBA contain stress-specific BFU-E that are more susceptible to TGF-β has suggested that galunisertib, an inhibitor of TGF-β receptor 1 signaling under clinical evaluation for a variety of diseases [73], may be an effective therapy for DBA [50].
In an effort to identify novel therapies for DBA, several animal models have been generated. These models are represented by zebrafish, drosophila and mouse strains genetically modified to carry mutations in ribosomal genes similar to those identified in patients [74]. These models have been instrumental to detail the defective ribosome biogenesis induced by DBA mutations [74]. However, since their phenotype includes only a modest macrocytic anemia which does not require transfusion, they are not recommended for the identification of novel strategies to cure the disease. The search for novel treatment strategies for this disease is therefore still an active ongoing process that heavily relays on the use of in vitro models of the disease and that may be facilitated by critical analysis of the confounding factors present in these models.
Interaction between signaling from cKIT, the receptor for SCF, and from GR
The first insights on the molecular abnormalities of DBA using ex vivo expanded cells were published by Horos et al. in 2012 [75]. By using cultures stimulated with murine SCF, EPO and Dex (Table 1), this study confirmed that CD34+ cells from some but not all DBA patients generate in culture good numbers of erythroid cells [70] and identified that the mutations in genes encoding ribosomal proteins (RP) associated with DBA impair mRNA binding to 40S, providing a mechanism which may reduce mRNA translation in these cells. The recognition that GR activation inhibits mRNA binding to ribosomes in muscle cells [62–66] is a clear indication that the presence of Dex in these cultures represents a confounding factor. However, the cultures contained murine SCF which is structurally similar (83% identical by BLAST search) [76] to the human growth factor but unable to activate the SCF-dependent ERK signaling that protects GR from lysosomal degradation in human erythroid cells [77]. Therefore, human erythroid cells grown with murine SCF are unlikely to contain levels of GR sufficiently high to elicit a Dex response. Whether synergy between cKIT and GR signaling underlies the improvements in erythroid differentiation observed in cultures of DBA stimulated with SCF [69–71] remains an open question.
Table 1. Summary of the culture conditions and expression results in four of the DBA studies published so far.
In addition to the differences reported by Ulirsch et al [10], some of which are summarized in black fonts, the four studies adopted greatly different improved culture conditions wired with novel confounding factors discussed in this commentary (in red fonts). NA=not applicable. Modified from [10].
Horos et al.75 | Ludwig et at.78 | Yang et al.89 | O’Brien et at.79 | |
---|---|---|---|---|
| ||||
Sample type | Peripheral blood | Bone marrow | Bone marrow | CD34+ peripheral blood |
| ||||
Sample origin | DBA (RPS19, RPL11) (n=11) |
![]() |
![]() |
![]() |
Controls (n=5) |
![]() |
![]() |
![]() |
|
| ||||
Patient growth | 7/11 | 3/3 | 3/3 | 7/11 |
| ||||
Culture | One step | One step | Three steps | Two steps (7 + 7 days) |
| ||||
Step 1 | ||||
Growth Factors | EPO (0.5U/mL), murine-SCF (100ng/mL) | SCF(10ng/mL), IL-3(1ng/mL), EPO (3U/mL) | SCF(100ng/mL), IL-3(5ng/mL), EPO (2U/mL) | SCF (100ng/mL), IL-3 (10ng/mL), EPO (0.5U/mL), IL-6 (10ng/mL), TPO (50ng/mL) |
Glucocorticoids | + Dex (10−6M) | + Dex (2×10−6M) | + HC (10−6M) | None |
| ||||
Step 2 | ||||
Growth Factors | NA | NA | SCF (100ng/mL), EPO (2U/mL) | EPO (3U/mL) |
| ||||
Step 3 | ||||
Growth Factors | NA | NA | EPO (2U/mL) | NA |
| ||||
Cells Analyzed | Day 16 | Primary Marrow Cells | Multiple, up to 17 days | Day 14 |
| ||||
Purification | None | CD34+CD71+ CD45+ | CD71+CD36−CD235a− CD71+CD36+CD235a+ CD36+CD235a+CD36−CD235a+ |
![]() |
| ||||
Platform | RT-qPCR | Affimetrix U133A | RT-qPCR | Affimetrix Human Gene 2.0 ST |
| ||||
ALAS2 mRNA | Not done | No change | Decreased | Decreased |
| ||||
GATA1 mRNA | Not done | No change | Not measured | No change |
| ||||
β-globin mRNA | Not done | No change | Decreased | Increased |
| ||||
GATA1 targets | Not done | Decreased | N/A |
![]() |
| ||||
Heme biosynthesis | Not done | Decreased | N/A |
![]() |
| ||||
Ribosome biogenesis | Decreased | Decreased | N/A | Decreased |
The observation that RP-deficiency prevents mRNA binding to ribosomes was confirmed in 2014 by Ludwig et al. [78] (Table 1). This group also reported that impairment is restricted to a subset of erythroid mRNA which include that encoding the long form of the transcription factor GATA1 which is specifically transcribed at late stages of maturation [78]. By identifying a DBA patient carrying a loss-of-function GATA1 mutation and by performing RP loss-of-function and GATA1 gain-of-function experiments with primary mobilized CD34+ cells, this study provided compelling evidence that reduced translation of GATA1 mRNA is responsible for differentiation abortion and insufficient red cell production in DBA. However, the cultures used in this study were stimulated with Dex in combination with human SCF, which assures that GR is optimally expressed. Therefore, these results are biased by the possibility that inhibition of mRNA binding to the ribosomes was, at least in part, the consequence of GR activation [62–66]. This question was finally clarified in a non-confounding fashion by O’Brien et al. who, by using cultures not supplemented with Dex, confirmed that erythroid cells derived from DBA patients carrying RP mutations express an abnormal ribosomal signature [79] (Table 1). Furthermore, this paper identified that similar ribosomal abnormalities are detected in cells generated from patients carrying the GATA1 mutation establishing a feed-back loop between GATA1 and the control of its mRNA translation. This provocative conclusion is independently supported by the observation that the hypomorphic Gata1 mutation induces abnormal RSP14 signature and deficient ribosome biogenesis in mice [80].
The fact that these three studies reached the same conclusion regardless of the presence of Dex makes questionable whether GR exerts the same effects on ribosome biogenesis in erythroid and muscle cells. Since GR agonists improve red blood cell production at least in some DBA patients [67,68], it is possible that, by contrast with muscle cells, in erythroid cells GR agonists elicit a non-canonical GR signal which improves ribosome biogenesis. Surprisingly in view of their obvious significance for ribosomopathies, the effects of these drugs on mRNA translation in erythroid cells have not been investigated as yet. Because of their clear clinical relevance, these effects and how they are affected by the regulation exerted by SCF on GR stability deserve to be carefully investigated. These studies may be relevant not only for DBA but also for other ribosomopathies. In fact, human GR is encoded by NR3C1 located on the long arms of chromosome 5 (5q31) close to the breakpoint of the chromosomal del(5q) abnormality deleted in patients with myelodisplastic syndrome (MDS), a disease also associated with anemia due to mutations in RP genes and reduced ribosome functions. Patients with del(5q) MDS often present with EPO-resistant anemia. A retrospective FISH analyses for NR3C1 in 14 of these patients identified that in 78% of the cases the breakpoint involved GR suggesting that GR haploinsufficiency exacerbates the ribosomal dysfunctions observed in these patients [81].
Patient treatment state
O’Brien et al. confirmed that CD34+ cells from only a sub-group of DBA patients generate good numbers of erythroid cells in culture and made the original important observation that CD34+ cells from glucocorticoid-responsive patients generate greater numbers of erythroid cells when collected after treatment [79]. This observation suggests that the responsive- and treatment-state of the patients are confounding factors when assessing the growth of DBA patients in culture. In addition, the fact that the cultures did not contain Dex allows speculating that the “greater expansions” observed with in vivo treated cells reflect the frequency in these populations of stress-specific erythroid progenitor cells [50] which may have been increased by glucocorticoids by promoting both commitment from earlier compartments, thanks to their chromatin remodeling effects [82], and self-renewal [43]. This observation is a paradigm shift in our understanding of the pathology and cure of DBA because it moves the abnormalities induced by the causative mutations and the therapeutic effects exerted by glucocorticoids up to the stem cell level, possibly at the level of the bipotent erythroid-megakaryocytic stem cell recently identify by Velten et al [83]. Since erythroid-megakaryocytic stem cells are extremely sensitive to thrombopoietin, this hypothesis deserves to be further investigated because it suggests that DBA patients may be responsive to the advanced MPL agonists, such as Eltrombopag, which have been proved effective in MDS and in aplastic anemia [84,85].
Cell proliferation state
Ludwig et al. reported that primary stem/progenitor cells (CD34pos) from DBA patients express normal levels of 5′-Aminolevulinate Synthase 2 (ALAS2, the first protein of the heme biosynthetic pathway) mRNA and decreased levels of mRNA encoding other proteins involved in heme biogenesis [78] (Table 1). By contrast, O’Brien et al. reported that the levels of ALAS2 and of other mRNA involved in heme biosynthesis are respectively decreased and increased in early erythroid cells (CD44+CD235a−) generated in vitro from DBA patients [79]. The difference between the results reported by the two groups may relate to the different populations analyzed (stem/progenitor cells vs early erythroid cells) and, as discussed by Ulirsch et al. [10], to the poor definition of stage maturation by O’Brian et al. In fact O’Brian et al purified their cells on the basis of CD44 expression, a good marker for the maturation of murine erythroid cells [86] but not for that of the human ones [49]. However, it is also possible that the differences are related to the proliferation state of the cells analyzed. In fact, in human erythroid cells, as in normal and leukemic stem cells [87], CD44 expression predicts proliferation potential [46]. Since post-natal CD34pos cells are mostly quiescent while CD44+ erythroid cells are all in cycle [46], it is possible that differences in mRNA levels observed among cell populations isolated with the CD44 marker highlight abnormalities affecting the ability of the cells to proliferate. This hypothesis is consistent with results published by the Abkowitz laboratory that, by defining erythroid maturation using the canonical CD71, CD36 and CD235a markers [88], confirmed that the most immature, and therefore with greater proliferation potential, erythroid cells expanded from DBA are those that express reduced levels of ALAS2 mRNA [89] (Table 1). However, consistently with the greater levels of mRNA encoding proteins of the heme byosythetic pathway reported by O’Brian et al [79], this study also reported that these cells contain increased levels of ALAS2 protein and of heme and that increased levels of heme are responsible for accelerated cell death [89]. The identification of cell proliferation potential as confounding factor is related to the fact that, by contrast with murine erythropoiesis in which maturation progresses regularly along four terminal divisions, in human cells terminal maturation and loss of proliferation potential are partially uncoupled. In fact, murine CFU-E-derived colonies contain up to 32–64 cells (i.e. 5–6 divisions) while the corresponding human colonies contain as many as 100–200 cells (> 7 divisions). The plasticity in the speed with which human erythroid cells lose proliferation potential with maturation may be related to species-specific differences in the speed of recovery from anemia and is also a clinically relevant area of research which deserves to be further investigated.
Conclusion
In addition to “maturation state” and other parameters already identified, ex vivo modeling of human erythropoiesis is prone to new “confounding factors” which may challenge the comparison of data obtained by different laboratories but, as already demonstrated in the hemoglobin switching field, may also unveil novel clinically relevant biological phenomena. In the case of DBA, and possibly of other diseases associated with ribosomopathy, systematic analyses of these confounding factors may provide hints on the mechanisms which lead to disease manifestation, glucocorticoids response and identify novel therapeutic targets.
Supplementary Material
Time lapse observations of erythroblasts and macrophages obtained at day 10 in culture of polycythemia vera. Erythroblasts and macrophages can be recognized by size. This movie reveals that the erythroid island may exhibit a highly dynamic nature as indicated by the numerous interactions occurring among erythroblasts and among 1–2 erythroblast(s) (indicated by the circle) and one macrophage (indicated by the arrow). The interaction between the erythroblasts and the macrophage is “loose” and lasts for at least one hour. After leaving the macrophage, the two erythroblasts (indicated by the circle) undergo cytokinesis. In addition to loose interactions, erythroblasts and macrophages may engage in “tight” interactions which support enucleation (see 92 for further details). A field at low cell density is shown for clarity but similar interactions are observed in areas of the dish at high cell density. Images captured with 10X objective each 30″ for about 1 hour and 40 minutes; reproduction speed at 8 frames per second.
Significance Statement.
By contrast with theoretical science, experimental science tests models which by definition are imperfect because they contain “confounding factors”. In the past, confounding factors identified in the in vitro models developed to study human erythropoiesis has greatly contributed to advance the field. Recent technological advances have greatly improved the power of analyses provided by in vitro models of human erythropoiesis. However, these improved models contain novel “confounding factors” which have yet to be fully discussed. This review will illustrate three novel confounding factors wired in advanced in vitro models of human erythropoiesis and discuss how their identification may drive additional studies to improve our understanding of the pathobiology of Diamond Blackfan Anemia and possibly other erythroid disorders.
Acknowledgments
This study was supported by grants from the National Cancer Institute (P01-CA108671) and Associazione Italiana Ricerca Cancro (AIRC 17608). Dr. Mario Falchi, Istituto Superiore di Sanita’, Rome, Italy, is gratefully acknowledged for providing the confocal microscopy observation and the movie of erythroid cells generated in culture.
Footnotes
Authors’ contribution
ARM and LV reviewed the literature and wrote the manuscript. All the authors have read the manuscript, concur with its content and state that its content has not been submitted elsewhere.
Conflict of interest disclosure
ARM and LV declare no competing financial interest.
References
- 1.Metcalf D. Hemopoietic colonies: in vitro cloning of normal and leukemic cells. Recent Results Cancer Res. 1977;61:1–227. [PubMed] [Google Scholar]
- 2.Metcalf D. The colony-stimulating factors and cancer. Cancer Immunol Res. 2013;1:351–6. doi: 10.1158/2326-6066.CIR-13-0151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Prchal JF, Adamson JW, Murphy S, Steinmann L, Fialkow PJ. Polycythemia vera. The in vitro response of normal and abnormal stem cell lines to erythropoietin. J Clin Invest. 1978;61:1044–7. doi: 10.1172/JCI109003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Miyake T, Kung CK, Goldwasser E. Purification of human erythropoietin. Biol Chem. 1977 Aug 10;252(15):5558–64. [PubMed] [Google Scholar]
- 5.Huisman TH, Reese AL, Gravely ME, Wilson JB, Webber B, Felice AE. Adult and fetal hemoglobin production in erythroid colonies from subjects with beta-thalassemia or with hereditary persistance of fetal hemoglobin (HPFH) Hemoglobin. 1980;4:449–67. doi: 10.3109/03630268008996226. [DOI] [PubMed] [Google Scholar]
- 6.Papayannopoulou T, Nakamoto B, Buckley J, Kurachi S, Nute PE, Stamatoyannopoulos G. Erythroid progenitors circulating in the blood of adult individuals produce fetal hemoglobin in culture. Science. 1978;199:1349–50. doi: 10.1126/science.628844. [DOI] [PubMed] [Google Scholar]
- 7.Papayannopoulou TH, Brice M, Stamatoyannopoulos G. Stimulation of fetal hemoglobin synthesis in bone marrow cultures from adult individuals. Proc Natl Acad Sci U S A. 1976;73(6):2033–2037. doi: 10.1073/pnas.73.6.2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kidoguchi K, Ogawa M, Karam JD, Martin AG. Augmentation of fetal hemoglobin (HbF) synthesis in culture by human erythropoietic precursors in the marrow and peripheral blood: studies in sickle cell anemia and nonhemoglobinopathic adults. Blood. 1978;52(6):1115–1124. [PubMed] [Google Scholar]
- 9.Peschle C, Migliaccio G, Covelli A, et al. Hemoglobin synthesis in individual bursts from normal adult blood: all bursts and subcolonies synthesize G gamma-and A gamma-globin chains. Blood. 1980;56(2):218–226. [PubMed] [Google Scholar]
- 10.Ulirsch JC, Lareau C, Ludwig LS, Mohandas N, Nathan DG, Sankaran VG. Confounding in ex vivo models of Diamond-Blackfan anemia. Blood. 2017 doi: 10.1182/blood-2017-05-783191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Papayannopoulou T, Kalmantis T, Stamatoyannopoulos G. Cellular regulation of hemoglobin switching: evidence for inverse relationship between fetal hemoglobin synthesis and degree of maturity of human erythroid cells. Proc Natl Acad Sci U S A. 1979;76(12):6420–6424. doi: 10.1073/pnas.76.12.6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chui DH, Wong SC, Enkin MW, Patterson M, Ives RA. Proportion of fetal hemoglobin synthesis decreases during erythroid cell maturation. Proc Natl Acad Sci U S A. 1980;77(5):2757–2761. doi: 10.1073/pnas.77.5.2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gianni AM, Comi P, Giglioni B, et al. Biosynthesis of Hb in individual fetal liver bursts. gamma-Chain production peaks earlier than beta-chain in the erythropoietic pathway. Exp Cell Res. 1980;130(2):345–352. doi: 10.1016/0014-4827(80)90011-7. [DOI] [PubMed] [Google Scholar]
- 14.Dover GJ, Boyer SH. Quantitation of hemoglobins within individual red cells: asynchronous biosynthesis of fetal and adult hemoglobin during erythroid maturation in normal subjects. Blood. 1980;56(6):1082–1091. [PubMed] [Google Scholar]
- 15.Papayannopoulou T, Kurachi S, Nakamoto B, Zanjani ED, Stamatoyannopoulos G. Hemoglobin switching in culture: evidence for a humoral factor that induces switching in adult and neonatal but not fetal erythroid cells. Proc Natl Acad Sci U S A. 1982;79(21):6579–6583. doi: 10.1073/pnas.79.21.6579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Constantoulakis P, Nakamoto B, Papayannopoulou T, Stamatoyannopoulos G. Fetal calf serum contains activities that induce fetal hemoglobin in adult erythroid cell cultures. Blood. 1990;75(9):1862–1869. [PubMed] [Google Scholar]
- 17.Migliaccio AR, Migliaccio G, Brice M, Constantoulakis P, Stamatoyannopoulos G, Papayannopoulou T. Influence of recombinant hematopoietins and of fetal bovine serum on the globin synthetic pattern of human BFUe. Blood. 1990;76(6):1150–1157. [PubMed] [Google Scholar]
- 18.Telen MJ. Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood. 2016;127(7):810–819. doi: 10.1182/blood-2015-09-618553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Belay E, Hayes BJ, Blau CA, Torok-Storb B. Human Cord Blood and Bone Marrow CD34+ Cells Generate Macrophages That Support Erythroid Islands. PLoS One. 2017;12(1):e0171096. doi: 10.1371/journal.pone.0171096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chow A, Huggins M, Ahmed J, et al. CD169(+) macrophages provide a niche promoting erythropoiesis under homeostasis and stress. Nat Med. 2013;19(4):429–436. doi: 10.1038/nm.3057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Soni S, Bala S, Gwynn B, Sahr KE, Peters LL, Hanspal M. Absence of erythroblast macrophage protein (Emp) leads to failure of erythroblast nuclear extrusion. J Biol Chem. 2006;281(29):20181–20189. doi: 10.1074/jbc.M603226200. [DOI] [PubMed] [Google Scholar]
- 22.Pace BS, Liu L, Li B, Makala LH. Cell signaling pathways involved in drug-mediated fetal hemoglobin induction: Strategies to treat sickle cell disease. Exp Biol Med (Maywood) 2015;240(8):1050–1064. doi: 10.1177/1535370215596859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sripichai O, Kiefer CM, Bhanu NV, Tanno T, Noh SJ, Goh SH, Russell JE, Rognerud CL, Ou CN, Oneal PA, Meier ER, Gantt NM, Byrnes C, Lee YT, Dean A, Miller JL. Cytokine-mediated increases in fetal hemoglobin are associated with globin gene histone modification and transcription factor reprogramming. Blood. 2009;114:2299–306. doi: 10.1182/blood-2009-05-219386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Palstra RJ, de Laat W, Grosveld F. Beta-globin regulation and long-range interactions. Adv Genet. 2008;61:107–142. doi: 10.1016/S0065-2660(07)00004-1. [DOI] [PubMed] [Google Scholar]
- 25.Li Q, Peterson KR, Fang X, Stamatoyannopoulos G. Locus control regions. Blood. 2002;100(9):3077–3086. doi: 10.1182/blood-2002-04-1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Williams DE, Eisenman J, Baird A, et al. Identification of a ligand for the c-kit proto-oncogene. Cell. 1990;63(1):167–174. doi: 10.1016/0092-8674(90)90297-r. [DOI] [PubMed] [Google Scholar]
- 27.Zsebo KM, Wypych J, McNiece IK, et al. Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver--conditioned medium. Cell. 1990;63(1):195–201. doi: 10.1016/0092-8674(90)90300-4. [DOI] [PubMed] [Google Scholar]
- 28.Huang E, Nocka K, Beier DR, et al. The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus. Cell. 1990;63(1):225–233. doi: 10.1016/0092-8674(90)90303-v. [DOI] [PubMed] [Google Scholar]
- 29.Munugalavadla V, Kapur R. Role of c-Kit and erythropoietin receptor in erythropoiesis. Crit Rev Oncol Hematol. 2005;54(1):63–75. doi: 10.1016/j.critrevonc.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 30.Ziegler BL, Muller R, Valtieri M, et al. Unicellular-unilineage erythropoietic cultures: molecular analysis of regulatory gene expression at sibling cell level. Blood. 1999;93(10):3355–3368. [PubMed] [Google Scholar]
- 31.Migliaccio G, Migliaccio AR, Druzin ML, Giardina PJ, Zsebo KM, Adamson JW. Long-term generation of colony-forming cells in liquid culture of CD34+ cord blood cells in the presence of recombinant human stem cell factor. Blood. 1992;79(10):2620–2627. [PubMed] [Google Scholar]
- 32.Miyazawa K, Williams DA, Gotoh A, Nishimaki J, Broxmeyer HE, Toyama K. Membrane-bound Steel factor induces more persistent tyrosine kinase activation and longer life span of c-kit gene-encoded protein than its soluble form. Blood. 1995;85(3):641–649. [PubMed] [Google Scholar]
- 33.Mattiucci D, Maurizi G, Izzi V, et al. Bone marrow adipocytes support haematopoietic stem cell survival. J Cell Physiol. 2017 doi: 10.1002/jcp.26037. [DOI] [PubMed] [Google Scholar]
- 34.Schofield KP, Humphries MJ, de Wynter E, Testa N, Gallagher JT. The effect of alpha4 beta1-integrin binding sequences of fibronectin on growth of cells from human hematopoietic progenitors. Blood. 1998;91(9):3230–3238. [PubMed] [Google Scholar]
- 35.Krystal G. A simple microassay for erythropoietin based on 3H-thymidine incorporation into spleen cells from phenylhydrazine treated mice. Exp Hematol. 1983;11(7):649–660. [PubMed] [Google Scholar]
- 36.Erslev AJ, Wilson J, Caro J. Erythropoietin titers in anemic, nonuremic patients. J Lab Clin Med. 1987;109(4):429–433. [PubMed] [Google Scholar]
- 37.Bauer A, Tronche F, Wessely O, et al. The glucocorticoid receptor is required for stress erythropoiesis. Genes Dev. 1999;13(22):2996–3002. doi: 10.1101/gad.13.22.2996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gursoy A, Dogruk Unal A, Ayturk S, et al. Polycythemia as the first manifestation of Cushing’s disease. J Endocrinol Invest. 2006;29(8):742–744. doi: 10.1007/BF03344186. [DOI] [PubMed] [Google Scholar]
- 39.Ellis H. Thomas Addison: Addisonian (pernicious) anaemia, Addison’s disease of the suprarenal gland. J Perioper Pract. 2013;23(1–2):31–32. doi: 10.1177/1750458913023001-205. [DOI] [PubMed] [Google Scholar]
- 40.Fibach E, Rachmilewitz EA. The two-step liquid culture: a novel procedure for studying maturation of human normal and pathological erythroid precursors. Stem Cells. 1993;11(Suppl 1):36–41. doi: 10.1002/stem.5530110608. [DOI] [PubMed] [Google Scholar]
- 41.Migliaccio G, Di Pietro R, di Giacomo V, et al. In vitro mass production of human erythroid cells from the blood of normal donors and of thalassemic patients. Blood Cells Mol Dis. 2002;28(2):169–180. doi: 10.1006/bcmd.2002.0502. [DOI] [PubMed] [Google Scholar]
- 42.von Lindern M, Zauner W, Mellitzer G, et al. The glucocorticoid receptor cooperates with the erythropoietin receptor and c-Kit to enhance and sustain proliferation of erythroid progenitors in vitro. Blood. 1999;94(2):550–559. [PubMed] [Google Scholar]
- 43.Lee HY, Gao X, Barrasa MI, et al. PPAR-alpha and glucocorticoid receptor synergize to promote erythroid progenitor self-renewal. Nature. 2015;522(7557):474–477. doi: 10.1038/nature14326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhang L, Prak L, Rayon-Estrada V, et al. ZFP36L2 is required for self-renewal of early burst-forming unit erythroid progenitors. Nature. 2013;499(7456):92–96. doi: 10.1038/nature12215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Paulson RF, Shi L, Wu DC. Stress erythropoiesis: new signals and new stress progenitor cells. Curr Opin Hematol. 2011;18(3):139–145. doi: 10.1097/MOH.0b013e32834521c8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Migliaccio G, Masiello F, Tirelli V, et al. Under HEMA conditions, self-replication of human erythroblasts is limited by autophagic death. Blood Cells Mol Dis. 2011;47(3):182–197. doi: 10.1016/j.bcmd.2011.06.001. [DOI] [PubMed] [Google Scholar]
- 47.Kolbus A, Blazquez-Domingo M, Carotta S, et al. Cooperative signaling between cytokine receptors and the glucocorticoid receptor in the expansion of erythroid progenitors: molecular analysis by expression profiling. Blood. 2003;102(9):3136–3146. doi: 10.1182/blood-2003-03-0923. [DOI] [PubMed] [Google Scholar]
- 48.Ghinassi B, Ferro L, Masiello F, et al. Recovery and Biodistribution of Ex Vivo Expanded Human Erythroblasts Injected into NOD/SCID/IL2Rgamma mice. Stem Cells Int. 2011;2011:673752. doi: 10.4061/2011/673752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hu J, Liu J, Xue F, et al. Isolation and functional characterization of human erythroblasts at distinct stages: implications for understanding of normal and disordered erythropoiesis in vivo. Blood. 2013;121(16):3246–3253. doi: 10.1182/blood-2013-01-476390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gao X, Lee HY, da Rocha EL, et al. TGF-beta inhibitors stimulate red blood cell production by enhancing self-renewal of BFU-E erythroid progenitors. Blood. 2016;128(23):2637–2641. doi: 10.1182/blood-2016-05-718320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Perry JM, Harandi OF, Paulson RF. BMP4, SCF, and hypoxia cooperatively regulate the expansion of murine stress erythroid progenitors. Blood. 2007;109(10):4494–4502. doi: 10.1182/blood-2006-04-016154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Xiang J, Wu DC, Chen Y, Paulson RF. In vitro culture of stress erythroid progenitors identifies distinct progenitor populations and analogous human progenitors. Blood. 2015;125(11):1803–1812. doi: 10.1182/blood-2014-07-591453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Leberbauer C, Boulme F, Unfried G, Huber J, Beug H, Mullner EW. Different steroids co-regulate long-term expansion versus terminal differentiation in primary human erythroid progenitors. Blood. 2005;105(1):85–94. doi: 10.1182/blood-2004-03-1002. [DOI] [PubMed] [Google Scholar]
- 54.Huang J, Liu X, Li D, et al. Dynamic Control of Enhancer Repertoires Drives Lineage and Stage-Specific Transcription during Hematopoiesis. Dev Cell. 2016;36(1):9–23. doi: 10.1016/j.devcel.2015.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.McKinney-Freeman S, Cahan P, Li H, et al. The transcriptional landscape of hematopoietic stem cell ontogeny. Cell Stem Cell. 2012;11(5):701–714. doi: 10.1016/j.stem.2012.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.An X, Schulz VP, Mohandas N, Gallagher PG. Human and murine erythropoiesis. Curr Opin Hematol. 2015;22(3):206–211. doi: 10.1097/MOH.0000000000000134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Migliaccio G, Migliaccio AR, Adamson JW. In vitro differentiation of human granulocyte/macrophage and erythroid progenitors: comparative analysis of the influence of recombinant human erythropoietin, G-CSF, GM-CSF, and IL-3 in serum-supplemented and serum-deprived cultures. Blood. 1988;72(1):248–256. [PubMed] [Google Scholar]
- 58.Verma R, Su S, McCrann DJ, et al. RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development. J Exp Med. 2014;211(9):1715–1722. doi: 10.1084/jem.20130624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schaefer A, Magocsi M, Marquardt H. Signalling mechanisms in erythropoiesis: the enigmatic role of calcium. Cell Signal. 1997;9(7):483–495. doi: 10.1016/s0898-6568(97)00094-6. [DOI] [PubMed] [Google Scholar]
- 60.Farrar JE, Neuberg D, Triche T, Bodine DM. Making “perfect” the enemy of good. Blood. 2017;(9):1168–1169. doi: 10.1182/blood-2017-07-789107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Singleton BK, Frayne J, Anstee DJ. Blood group phenotypes resulting from mutations in erythroid transcription factors. Curr Opin Hematol. 2012;19(6):486–493. doi: 10.1097/MOH.0b013e328358f92e. [DOI] [PubMed] [Google Scholar]
- 62.Showkat M, Beigh MA, Andrabi KI. mTOR Signaling in Protein Translation Regulation: Implications in Cancer Genesis and Therapeutic Interventions. Mol Biol Int. 2014;2014:686984. doi: 10.1155/2014/686984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang H, Kubica N, Ellisen LW, Jefferson LS, Kimball SR. Dexamethasone represses signaling through the mammalian target of rapamycin in muscle cells by enhancing expression of REDD1. J Biol Chem. 2006;281(51):39128–39134. doi: 10.1074/jbc.M610023200. [DOI] [PubMed] [Google Scholar]
- 64.Shah OJ, Kimball SR, Jefferson LS. Glucocorticoids abate p70(S6k) and eIF4E function in L6 skeletal myoblasts. Am J Physiol Endocrinol Metab. 2000;279(1):E74–82. doi: 10.1152/ajpendo.2000.279.1.E74. [DOI] [PubMed] [Google Scholar]
- 65.Shah OJ, Kimball SR, Jefferson LS. Among translational effectors, p70S6k is uniquely sensitive to inhibition by glucocorticoids. Biochem J. 2000;347(Pt 2):389–397. doi: 10.1042/0264-6021:3470389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Shah OJ, Kimball SR, Jefferson LS. Acute attenuation of translation initiation and protein synthesis by glucocorticoids in skeletal muscle. Am J Physiol Endocrinol Metab. 2000;278(1):E76–82. doi: 10.1152/ajpendo.2000.278.1.E76. [DOI] [PubMed] [Google Scholar]
- 67.Vlachos A, Muir E. How I treat Diamond-Blackfan anemia. Blood. 2010;116(19):3715–3723. doi: 10.1182/blood-2010-02-251090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dianzani I, Garelli E, Ramenghi U. Diamond-Blackfan anemia: a congenital defect in erythropoiesis. Haematologica. 1996;81(6):560–572. [PubMed] [Google Scholar]
- 69.Abkowitz JL, Sabo KM, Nakamoto B, et al. Diamond-blackfan anemia: in vitro response of erythroid progenitors to the ligand for c-kit. Blood. 1991;78(9):2198–2202. [PubMed] [Google Scholar]
- 70.Olivieri NF, Grunberger T, Ben-David Y, et al. Diamond-Blackfan anemia: heterogenous response of hematopoietic progenitor cells in vitro to the protein product of the steel locus. Blood. 1991;78(9):2211–2215. [PubMed] [Google Scholar]
- 71.Bagnara GP, Zauli G, Vitale L, et al. In vitro growth and regulation of bone marrow enriched CD34+ hematopoietic progenitors in Diamond-Blackfan anemia. Blood. 1991;78(9):2203–2210. [PubMed] [Google Scholar]
- 72.Bellet JS, Obadiah JM, Frothingham BM, Kurtzberg J, Grichnik JM. A patient with extensive stem cell factor-induced hyperpigmentation. Cutis. 2003;71(2):149–152. [PubMed] [Google Scholar]
- 73.de Gramont A, Faivre S, Raymond E. Novel TGF-beta inhibitors ready for prime time in onco-immunology. Oncoimmunology. 2017;6(1):e1257453. doi: 10.1080/2162402X.2016.1257453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.McGowan KA, Mason PJ. Animal models of Diamond Blackfan anemia. Semin Hematol. 2011;48(2):106–116. doi: 10.1053/j.seminhematol.2011.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Horos R, Ijspeert H, Pospisilova D, et al. Ribosomal deficiencies in Diamond-Blackfan anemia impair translation of transcripts essential for differentiation of murine and human erythroblasts. Blood. 2012;119(1):262–272. doi: 10.1182/blood-2011-06-358200. [DOI] [PubMed] [Google Scholar]
- 76.www.ncbi.nlm.nih.gov/gene?Db=gene&Cmd=DetailsSearch&Term=4254. 2016.
- 77.Varricchio L, Tirelli V, Masselli E, et al. The expression of the glucocorticoid receptor in human erythroblasts is uniquely regulated by KIT ligand: implications for stress erythropoiesis. Stem Cells Dev. 2012;21(15):2852–2865. doi: 10.1089/scd.2011.0676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ludwig LS, Gazda HT, Eng JC, et al. Altered translation of GATA1 in Diamond-Blackfan anemia. Nat Med. 2014;20(7):748–753. doi: 10.1038/nm.3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.O’Brien KA, Farrar JE, Vlachos A, et al. Molecular convergence in ex vivo models of Diamond-Blackfan anemia. Blood. 2017;129(23):3111–3120. doi: 10.1182/blood-2017-01-760462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zingariello M, Sancillo L, Martelli F, et al. The thrombopoietin/MPL axis is activated in the Gata1low mouse model of myelofibrosis and is associated with a defective RPS14 signature. Blood Cancer J. 2017;7(6):e572. doi: 10.1038/bcj.2017.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Varricchio L, Migliaccio AR. The Role of Glucocorticoid receptor (GR) polymorphisms in human erythropoiesis. Am J Blood Res. 2014;4:53–72. [PMC free article] [PubMed] [Google Scholar]
- 82.Guo B, Huang X, Cooper S, Broxmeyer HE. Glucocorticoid hormone-induced chromatin remodeling enhances human hematopoietic stem cell homing and engraftment. Nat Med. 2017;23(4):424–428. doi: 10.1038/nm.4298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Velten L, Haas SF, Raffel S, et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat Cell Biol. 2017;19(4):271–281. doi: 10.1038/ncb3493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Desmond R, Townsley DM, Dumitriu B, Olnes MJ, Scheinberg P, Bevans M, Parikh AR, Broder K, Calvo KR, Wu CO, Young NS, Dunbar CE. Eltrombopag restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on discontinuation of drug. Blood. 2014;123:1818–25. doi: 10.1182/blood-2013-10-534743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mavroudi I, Pyrovolaki K, Pavlaki K, Kozana A, Psyllaki M, Kalpadakis C, Pontikoglou C, Papadaki HA. Effect of the nonpeptide thrombopoietin receptor agonist eltrombopag on megakaryopoiesis of patients with lower risk myelodysplastic syndrome. Leuk Res. 2011;35:323–8. doi: 10.1016/j.leukres.2010.06.029. [DOI] [PubMed] [Google Scholar]
- 86.Liu J, Zhang J, Ginzburg Y, et al. Quantitative analysis of murine terminal erythroid differentiation in vivo: novel method to study normal and disordered erythropoiesis. Blood. 2013;121(8):e43–49. doi: 10.1182/blood-2012-09-456079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zoller M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front Immunol. 2015;6:235. doi: 10.3389/fimmu.2015.00235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Giarratana MC, Kobari L, Lapillonne H, et al. Ex vivo generation of fully mature human red blood cells from hematopoietic stem cells. Nat Biotechnol. 2005;23(1):69–74. doi: 10.1038/nbt1047. [DOI] [PubMed] [Google Scholar]
- 89.Yang Z, Keel SB, Shimamura A, et al. Delayed globin synthesis leads to excess heme and the macrocytic anemia of Diamond Blackfan anemia and del(5q) myelodysplastic syndrome. Sci Transl Med. 2016;8(338):338ra367. doi: 10.1126/scitranslmed.aaf3006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Imai M, Araki M, Komatsu N. Somatic mutations of calreticulin in myeloproliferative neoplasms. Int J Hematol. 2017;105(6):743–747. doi: 10.1007/s12185-017-2246-9. [DOI] [PubMed] [Google Scholar]
- 91.Siatecka M, Bieker JJ. The multifunctional role of EKLF/KLF1 during erythropoiesis. Blood. 2011;118(8):2044–2054. doi: 10.1182/blood-2011-03-331371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Falchi M, Varricchio L, Martelli F, et al. Dexamethasone targeted directly to macrophages induces macrophage niches that promote erythroid expansion. Haematologica. 2015;100(2):178–187. doi: 10.3324/haematol.2014.114405. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Time lapse observations of erythroblasts and macrophages obtained at day 10 in culture of polycythemia vera. Erythroblasts and macrophages can be recognized by size. This movie reveals that the erythroid island may exhibit a highly dynamic nature as indicated by the numerous interactions occurring among erythroblasts and among 1–2 erythroblast(s) (indicated by the circle) and one macrophage (indicated by the arrow). The interaction between the erythroblasts and the macrophage is “loose” and lasts for at least one hour. After leaving the macrophage, the two erythroblasts (indicated by the circle) undergo cytokinesis. In addition to loose interactions, erythroblasts and macrophages may engage in “tight” interactions which support enucleation (see 92 for further details). A field at low cell density is shown for clarity but similar interactions are observed in areas of the dish at high cell density. Images captured with 10X objective each 30″ for about 1 hour and 40 minutes; reproduction speed at 8 frames per second.