You are on page 1of 10

Erythroid cells in vitro: from developmental biology to blood

transfusion products
Anna Rita Migliaccioa,b, Carolyn Whitsetta,c and Giovanni Migliacciod
a
Tisch Cancer Institute, Mount Sinai School of Purpose of review
Medicine, bMyeloproliferative Research Consortium
(MPD-RC), cNew York Blood Center, New York, New
Red blood cells (RBCs) transfusion plays a critical role in numerous therapies.
York, USA and dDepartment of Cell Biology and Disruption of blood collection by political unrest, natural disasters and emerging
Neurosciences, Istituto Superiore Sanità, Rome, Italy
infections and implementation of restrictions on the use of erythropoiesis-stimulating
Correspondence to Professor Anna Rita Migliaccio, agents in cancer may impact blood availability in the near future. These considerations
PhD, Tisch Cancer Institute, Mount Sinai School of
Medicine, One Gustave L Levy Place, Box 1079, highlight the importance of developing alternative blood products.
New York, NY 10029, USA Recent findings
Tel: +1 212 2416974; fax: +1 212 8765276;
e-mail: annarita.migliaccio@mssm.edu Knowledge about the processes that control RBC production has been applied to the
establishment of culture conditions allowing ex-vivo generation of RBCs in numbers
Current Opinion in Hematology 2009,
16:259–268
close to those (2.5  1012 cells/ml) present in a transfusion, from cord blood, donated
blood units or embryonic stem cells. In addition, experimental studies demonstrate that
such cells protect mice from lethal bleeding. Therefore, erythroid cells generated ex vivo
may be suitable for transfusion provided they can be produced safely in adequate
numbers. However, much remains to be done to translate a theoretical production of
approximately 2.5  1012 RBCs in the laboratory into a ‘clinical grade production
process’.
Summary
This review summarizes the state-of-the-art in establishing ex-vivo culture conditions for
erythroid cells and discusses the most compelling issues to be addressed to translate
this progress into a clinical grade transfusion product.

Keywords
alternative blood transfusion products, cord blood, erythroid cells, erythropoietin,
ex-vivo expansion

Curr Opin Hematol 16:259–268


ß 2009 Wolters Kluwer Health | Lippincott Williams & Wilkins
1065-6251

ditions are common [3]. In emergencies, national plans


Introduction call for sharing of blood resources across geographic areas,
The safety and adequacy of the blood supply is a national accessing frozen blood inventories and sponsoring emer-
[1–4] and international (http://www.who.int/bloodsafety/ gency blood drives. However, the infrastructure to main-
universalbts/en/) [5] priority. In developed countries, the tain blood collection and distribution systems may be
blood supply appears to be adequate. For example, in disrupted during a disaster.
2007, blood collection and utilization data from the USA
indicate that the number of screened allogeneic blood Even when blood supplies are adequate, finding blood for
donations that passed all tests exceeded by 7.8% of the certain patients is difficult. Chronically transfused
total number of units transfused [1]. WHO reports, how- patients may become immunized to inherited poly-
ever, indicate that in developing countries national morphic cell surface determinants (minor blood group
supplies are rarely sufficient to meet existing needs antigens) [12] (Table 1). Clinicians often choose to
(http://www.who.int/bloodsafety/universalbts/en/) [5]. transfuse patients at high risk for alloimmunization with
Recent changes in health policies that restrict the use fully matched blood. Regional, national and international
of erythropoiesis-stimulating agents in patients with can- programs have been developed to identify blood for
cer [6,7] and reports of increased morbidity and mortality alloimmunized patients needing rare blood types [9].
associated with transfusion of older blood units (if con- National rare donor registries collaborate through the
firmed) [8] may decrease the blood supply in developed WHO International Rare Donor Panel to provide rare
countries in the future (Table 1) [9–13]. Sporadic blood blood wherever it is needed [9]. Despite unprecedented
shortages related to holidays and extreme weather con- efforts to meet the need of these special patients, there is
1065-6251 ß 2009 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI:10.1097/MOH.0b013e32832bcaa2

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
260 Hematopoiesis

Table 1 Factors that negatively affect the blood supply


Donor population Increased travel-related deferrals
Epidemics/emerging infections
Identification of new transfusion transmitted infections
Ethnically imbalanced donor/patient populations
Societal and cultural factors
Patient population Size
Limitation on the use of EPO
Increases in the minimal hematocrit for transfusion
Increases in the numbers of stem cell and solid-organ transplants
Lack of best transfusion practice guidelines
Innovative surgical treatments
Special needs
Neonates (fresher units preferred)
Increase in alloimmunized patientsa
Increasing demand for rare blood types and CMV-negative blood
Blood quality Product modifications that decrease shelf life (irradiation and washing)
Possible toxicity of older units may decrease inventory (current shelf life of blood 42 days)
Natural disasters Disrupt blood collection and manufacturing infrastructure
Interrupt communications between blood banks and local hospitals
Increase in patient population
Decrease in donor population
CMV, cytomegalovirus; EPO, erythropoietin. Data from [9–13].
a
In the USA, examples of programs dedicated to the identification of blood for alloimmunized patients include the Blue-tag Program in Philadelphia,
Precise Match in New York City (both regional programs) and the American Rare Donor Program (a national program). International efforts are
described in [9].

a shortage of certain rare blood types, hence the need to made in establishing ex-vivo culture conditions for ery-
identify alternative RBC transfusion products. throid cells and discuss the most compelling issues to be
addressed to translate this progress into a clinical grade
Frozen blood is the alternative transfusion product transfusion product.
usually provided for alloimmunized patients when liquid
units are not available [9]. Additional alternative trans-
fusion products not yet approved for clinical use in the Expansion of human erythroid cells in culture
USA or Europe are represented by hemoglobin (Hb) Erythroid differentiation begins at the level of the plur-
solutions and universal RBC transfusion products ipotent HSC [19]. HSCs are instructed by specific growth
(Table 2 [14,15]). RBCs generated in vitro from human factors to generate a hierarchy of progressively lineage-
hematopoietic stem/progenitor cells (HSCs) derived restricted erythroid progenitor cells (EPCs), a process
from cord [16] and adult [17] blood or human embryonic termed commitment. EPCs, morphologically indistin-
stem cells (hESCs) [18] may represent an important guishable from HSCs, are defined by specific antigen
resource for providing blood to patients with rare blood and mRNA expression profiles and give rise, after several
types and supplementing the general blood supply divisions, to the first morphologically recognizable ery-
during emergencies. We will review the recent progress throid cell, the proerythroblast. Proerythroblasts are still

Table 2 Red blood cell products and transfusion alternatives


Product/alternative Advantages Disadvantages

Frozen blood [9] Satisfactory storage life (>10 years) Not immediately available
Cell loss during deglycerolization
Expiration 24 h after thawing (14 days closed system)
Available only near blood centers and large hospitals
that have special facilities
Hb solutions [14] (recombinant/stabilized) Approved in South Africa for Not approved for human use in Europe or the USA
limited clinical use (approved in Europe for veterinary use only)
Short intravascular survival
Possible toxicity
Universal red cell transfusion products [15] Generates group O cells Currently not approved for clinical use
(enzymatic cleavage/chemical masking of Not applicable to all blood group antigens
ABO and Rh antigens)
EPO [6,7] Licensed Clinical indications limited
Expensive
Adverse reaction profile not defined for many
clinical conditions
EPO, erythropoietin; Hb, hemoglobin; Rh, rhesus.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Erythroid cells in vitro Migliaccio et al. 261

capable of proliferating and generating additional mature The two hormones act by delaying the transition of
erythroid precursors (basophilic, polychromatic and ortho- proerythroblasts to the next maturation stage, favoring
chromatic erythroblasts), which eventually lose prolifera- a higher number of proliferative events. Addition of
tive potential and undergo enucleation [20]. The number DXM and estradiol, alone or in combination, increases
of erythroblasts produced by this continuous passage the numbers of erythroblasts that can be obtained in
through distinct cellular compartments is determined both human cultures [16,17,27–32]. Cultures that increase
by the number of EPCs recruited and, more importantly, erythroblasts are defined as phase 1, expansion or human
by the number of divisions allowed to occur within each erythroid massive amplification (HEMA) cultures (Fig. 2
cell compartment preceding the polychromatic erythro- [17,27–33,34,35]). Calculation of the number of eryth-
blasts stage. roblasts that can be obtained from MNCs or CD34þ cells
from an average adult or cord blood unit under such
Polycythemia is the first manifestation of Cushing’s dis- conditions indicates that at least cord blood cells can
ease [21], a syndrome associated with chronic stimulation generate numbers of erythroblasts similar to those pre-
of the glucocorticoid receptor, whereas anemia is fre- sent in a transfusion (1.7  1011 vs. 2.5  1012) (Table 3
quently associated with estrogen therapy in breast cancer [36,37]).
[22]. These observations suggested that, in addition to
erythropoietin (EPO), erythropoiesis may also be As the purpose of expansion cultures is to optimize
regulated by nuclear receptors, such as glucocorticoid erythroblasts proliferation, a different set of conditions
receptor and the estrogen receptor (ESR), and prompted was designed to induce erythroblasts maturation (Fig. 2)
early studies that identified the ability of dexamethasone [16,17,27–29,30–32]. Ex-vivo expanded erythroblasts are
(DXM, a glucocorticoid receptor agonist) [23] and estra- transferred into maturation cultures, also referred to as
diol [24] to synergize with EPO in inducing generation of phase 2, stimulated with EPO and insulin-like growth
erythroid colonies in cultures of mononuclear cells factor (IGF)1. Under these conditions, erythroblasts pro-
(MNCs) from adult bone marrow or blood. Recently, gress to the orthochromatic stage (Fig. 3 [38,39]). Enu-
the role for these hormones in the regulation of hema- cleation remains inefficient (5–10%) and requires the
topoiesis has been confirmed by the observation that addition of MS-5 cells [29,33] that probably exert in vitro
mice lacking glucocorticoid receptor recover poorly from the functions performed by macrophages in vivo [40] (to
chemically induced anemia [25], a finding which led to provide a surface to facilitate rolling over during enuclea-
the discovery that addition of DXM to a combination of tion [41], to facilitate iron uptake, or both [42]) (Fig. 2).
growth factors used to stimulate murine EPCs results in Functionality studies proved that murine RBCs obtained
the generation of larger numbers of erythroblasts in in vitro from ESCs under these conditions protect mice
culture [26]. Now that recombinant growth factors are from lethal bleeding [43].
available, it is clear that DXM and estradiol do not affect
the number of EPCs recruited in the maturation process Given that EPCs represent a minority of CD34þ cells,
but rather the cellular output from each EPC (Fig. 1). culture conditions have been designed to induce CD34þ

Figure 1 Estradiol and dexamethasone do not increase the number of erythroid progenitor cells that form colonies in vitro but the
cellular output from each erythroid progenitor cell

Number and morphology of erythroid colonies obtained in culture of adult blood MNCs stimulated with optimal concentrations of SCF, IL-3 and EPO in
the presence of DXM, estradiol or both, as indicated. Magnification 10. DXM, dexamethasone; EPO, erythropoietin; IL, interleukin; MNCs,
mononuclear cells; SCF, stem cell factor.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
262 Hematopoiesis

Figure 2 Anatomy of a hypothetical ex-vivo red blood cell production process

Anatomy of an ex-vivo red blood cell production process

Preculture Phase 1 Phase 2


(commitment) (expansion) (maturation)

IL-3Rα IL-3Rβ GM-CSFRα IL-3Rβ GM-CSFRα EpoR EpoR


IL-3Rα IL-3Rα
EpoR EpoR
HSC EPC Red blood
EPC ProEBs ProEBs cells
gp130
gp130 Mp1 gp130 Mp1
Flt3 Kit Mp1 Kit Kit Mp1 Mp1

MS-5

Culture components Culture components Culture components

Starting cells: Cord blood (or adult blood) Starting cells: Cord blood or adult blood MNC; Starting cells: Erythroblasts obtained in
CD34+ cells cord blood or adult blood CD34+ cells; phase 1
Growth factors: FL; SCF; TPO; IL-6; GF-1 precultured of EPCs. Growth factors: EPO; IGF-1
Growth factors: SCF; IL-3/GM/CSF; EPO; IGF-1
Hormones: DXM; estradiol

Biomarkers Biomarkers Biomarkers

Potency of the donor: Genetic Quality control: Phenotypic (CD235a, CD36, Quality control: Phenotypic (CD235a, CD36,
polymorphisms, age, sex and ethnicity. CD71), or expression (GATA1 level) profiling; CD71), or expression (GATA1 level) profiling.
Potency of the EPC: Phenotypic profiling functionality assay (apoptosis).
(CD235a, CD36, CD71, others); gene Potency as transfusion product: Xenogeneic Potency as transfusion product:
expression profiling (GATA1/PU-1/or animal transfusion models. Xenogeneic animal transfusion models.
GATA1/GATA2 mRNA ratio), others;
Functional assays (colony formation).

Under development Transfusion product Transfusion product

Starting cells: hESC; hiPC. Advantages: Present in cord blood transfusions, Advantages: Large clinical experience.
easy to store and thaw; in-vivo maturation may
Epigenetic modifiers: HDAC inhibitors. deplete iron; longer efficacy; reduce the frequency Disadvantages: Glycerol dependent storage;
methylases inhibitors of transfusion in chronically transfused patients. the process of enucleation requires feeder
Disadvantages: Time required for efficacy not layers of murine origin; feeder layers of human
known; possible altered antigenicity; altered Hb origin are under development.
expression.

On the basis of the results obtained by our group [17] and others [27–29,30–32], a process to produce ex-vivo RBCs will be composed of three
sequential cultures: preculture (from HSCs to EPCs), expansion (from EPCs to erythroblasts) and maturation (from erythroblasts to enucleated RBCs).
cultures. Growth factor receptors expressed by the populations seeded in each culture and by their progeny are indicated. These expression patterns
guided the choice of growth factors used to stimulate each phase of the production process. The components (starting populations, growth factors and
additives) of each phase of the production are specified in blue boxes. Biomarkers suggested for quality control of cells generated in each culture are
indicated in green boxes. The pink boxes indicate advantages and disadvantages of using erythroblasts and enucleated RBCs as transfusion products
and the promising areas of research in the development of preculture systems. The very first process to generate ex-vivo RBC transfused products may
include only expansion cultures. It is envisioned that the first applications of ex-vivo generated RBC transfusion will be drug delivery [35] or possibly
transfusion of alloimmunized sickle cell patients, patients transplanted with cord blood stem cells, or both. These applications must be preceded by
proof of concept experiments in animal models. DXM, dexamethasone; EPO, erythropoietin; EPCs, erythroid progenitor cells; FBS, fetal bovine serum;
Hb, hemoglobin; HDAC, histone deacetylase; hESC, human embryonic stem cell; HSCs, hematopoietic stem cells; MNCs, mononuclear cells; RBC(s),
red blood cell(s); SCF, stem cell factor.

cells to generate EPCs that would, in turn, be used in chromatin, favoring and inhibiting, respectively, gene
expansion cultures. The purpose of these precultures/ expression. Once the chromatin configuration is estab-
commitment cultures is to increase the number of EPCs, lished, methylation of specific DNA regions ensures
and therefore of erythroblasts, that can be generated that the chromatin modeling is inherited by the cel-
ex vivo from a single blood donation. Preculture con- lular progeny. These plastic processes are regulated by
ditions have been developed for cord blood CD34þ cells families of enzymes called methylases, histone acetyl-
[32] but, in principle, the same conditions could be used transferases (HATs) and histone deacetylases (HDACs)
to generate EPCs from adult blood CD34þ cells (Fig. 2). [44]. The plasticity of this process has been recently
The transition from HSCs to EPCs is associated with exploited to reprogram mouse and human somatic cells
extensive chromatin remodeling that ensures activation by inducing ectopic expression of the four genes Oct4,
of the expression of erythroid-specific genes [44]. Sox2, Klf4 and cmyc (induced pluripotency cells or iPCs)
These epigenetic modifications involve acetylation/de- [45]. Exposure of murine fibroblasts to HDAC inhibi-
acetylation of specific histones that relaxes/condenses the tors increased 100-fold the otherwise low efficiency rate

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Erythroid cells in vitro Migliaccio et al. 263

Table 3 Calculation of the theoretical number of erythroid cells that would be generated by mononuclear cells and CD34R cells from
an average cord blood and adult blood unit cultured under expansion conditions
Adult blood Cord blood

Volume/unit (ml) 450–500 60–120


WBCs/unit (cells/ml) 4.5  109 9  108
HEMA culture seeded with MNCs
MNCs concentration (%) 10 10
Yield of the purification (%) 90–100 90–100
Total number of MNCs in culture (cells/ml) 4.3  108 8.6  107
Fold increase in culture 30 2000
Total number of erythroblasts obtained after 10–14 days of culture (cells/ml) 1.2  1010 1.7  1011
HEMA culture seeded with CD34þ cells

Total number of CD34þ cells per unit (cells/ml) 106( ) 1.8  106
CD34þ/MNC (%) 0.15 0.8
Yield of the purification (%) 50–100 50–100
Total number of CD34þ cells cultured (cells/ml) 4.8  105 6.6  105
Fold increase in culture 103 105
Total number of erythroblasts obtained after 10–14 days of culture (cells/ml) 4.8  108 6.6  1010
A unit of blood contains 2.5  1012 RBCs. The average frequencies of MNC and CD34þ cells in adult blood and cord blood are from [36,37]. The
frequencies of these cells in different units may vary by 1–2 logs. In spite of the 2-log increase obtained in cultures started with CD34þ cells, higher
numbers of erythroblasts are generated in cultures of MNCs. This is due to both loss of CD34þ cells during purification and to the fact that some EPCs
are CD34. EPCs, erythroid progenitor cells; HEMA, human erythroid massive amplification; MNC(s), mononuclear cell(s); RBCs, red blood cells;
WBCs, white blood cells.

of this reprogramming [46], suggesting that addition of is occasionally used for transfusions in developing
HDAC inhibitors to commitment cultures might allow the countries [47]. In 1918, Ende and Ende [48] reported
generation of higher numbers of EPCs as well (Fig. 2). that the majority of the RBCs circulating in a patient
transfused with 40 ml of cord blood were donor derived
for up to 2 months following the transfusion. This obser-
A clinical grade production process for vation and the consideration that erythroblasts may
ex-vivo generation of red blood cell undergo 4–64 further divisions suggest that ex-vivo
transfusion products generated erythroblasts may be more effective as trans-
In the USA, ex-vivo generated RBC transfusion products fusion products than regular transfusions (Table 4). Sup-
fall into the category of biological medicinal products. port for this hypothesis is provided by the fact that ex-
The identity of these products must be precisely docu- vivo generated erythroblasts, as those present in cord
mented by defining cell composition, nature of the pro- blood, express the adhesion receptors [chemokine (CXC
duction process and potency in xenogeneic animal motif) receptor 4, a4 integrin and P-selectin ligand],
models. In addition, internal controls must be devised necessary to establish proper cell interactions for matu-
to monitor the quality of cells at each stage of production ration once injected in vivo [49].
and the safety of the product. We will discuss further how
the knowledge of the biology of erythroid cells can guide The definition of the cellular composition of the product
the definition of an ex-vivo generated RBC transfusion must also take into account the fact that the biological
product. properties of erythroblasts change during ontogenesis
[20]. Therefore, erythroblasts generated from cord blood,
Cellular composition of the product adult blood, precultured cord blood or adult blood and
A conservative approach would be to define the cellular hESCs will likely express different biological properties.
composition of ex-vivo expanded RBC products as ‘enu- These differences include size, levels of activity of the
cleated RBCs’. The advantage of this definition is that glycolytic enzymes (higher in cord blood cells) and of
these cells are currently used for transfusion therapy. carbonic anhydrase (higher in adult blood cells), expres-
However, the high costs, long production times and sion of different isozymes (phosphoglycerate kinase,
presence of xenogeneic material (Fig. 2 and Table 4) acetylcholinesterase, etc.), HbF or HbA and antigenic
decrease the likelihood that this product will be produced profiles (human leukocyte antigen class II antigens may
under clinical grade conditions in the near future. be expressed by cord blood but not adult blood-derived
erythroblasts, cord blood or adult blood erythroblasts may
In principle, ex-vivo generated erythroblasts could also express the i or I antigen).
serve as the transfusion product. These cells are present
in the circulation during fetal development, in neonates Nature of the production process
with pyruvate kinase deficiency and in adults, under The nature of a production process is usually discussed in
conditions of stress [20]. Erythroblasts are also present terms of vessel types, incubation conditions, cell manip-
in large numbers (4  107 cells/ml) in cord blood, which ulation/collection/storage, and so on. All these issues are

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
264 Hematopoiesis

Figure 3 Biological properties of ex-vivo generated human erythroblasts

(a) Antigenic profiling of erythroblasts obtained after 10 days in expansion culture and of the progeny generated by these erythroblasts upon exposure
for 4 days to EPO in maturation cultures. Expansion cultures were seeded with adult blood MNCs and stimulated with SCF, IL-3, EPO, DXM and
estradiol. These cultures generated almost pure (>90%) erythroblasts, as demonstrated by CD36/CD235a staining. CD235a recognizes glycophorin
A. CD36 represents an early marker for erythroid commitment and recognizes the thrombospondin receptor, an antigen acquired by those
hematopoietic cells that had became EPCs in response to EPO [38]. Double CD36/CD235a staining divides erythroblasts into four populations:
EPCs (CD36high/CD235a), proerythroblasts (CD36high/CD235alow), basophilic–polychromatophilic (CD36high/CD235ahigh, basoerythroblast) and
orthochromatic (CD36/CD235ahigh, orthoerythroblast) erythroblasts [39]. Reticulocytes are identified as small CD36/CD235ahigh cells that stain
negative for DNA. The gatings identifying the four populations are indicated by rectangles. Erythroblasts obtained after 10 days of expansion cultures
are mainly composed of proerythroblasts and basoerythroblass. Removal of basoerythroblasts from the culture by density selection allows generation
of at least 20-fold more erythroblasts because these cells continue to proliferate up to day 25 of culture [28]. This observation suggests a strategy to
reduce the volume of the media required to produce an ex-vivo RBC transfusion product by sequential depletion/storage of basoerythroblasts.
Erythroblasts obtained in expansion cultures progress through the stage of orthochromatic erythroblast in 3–4 days when transferred into maturation
cultures stimulated with EPO and IGF-1 (or insulin). The low (5–18% depending on the donor) frequency of erythroblasts that will undergo enucleation
in these cultures can be greatly enhanced by the presence of the murine MS-5 cell line [29,33]. (b) FSC analyses of cells at different stages of
maturation indicating the significant size reduction associated with maturation of erythroblasts. Each curve corresponds to a different population, as
indicated on the right. Proerythroblasts (in red) were obtained in expansion cultures, whereas basoerythroblasts and orthoerythroblasts and RBCs were
at day 4 of maturation. (c) Time in culture and changes in morphology and size of human erythroblasts as they mature. The length of time and the
changes in size are compared with those observed during the maturation of adult erythroblasts in the marrow. Cell sizes are expressed as cell diameter
and were determined by comparison of the FSCs of the different populations presented in (b) with those of commercial beads of defined size. Ex-vivo
generated proerythroblasts range in size from 30 to 50 mm. These cells significantly reduce their diameter upon exposure to EPO from 40.1  1.4 to
28.5  2.0, 21.2  0.7 and 11.6  0.3 mm by 24, 48 and 96 h (P < 0.01, in all cases). Transition during the early phases of maturation occurs more
rapidly in vitro than in vivo, although later on maturation in vitro appears delayed (possibly because of absence of MS-5 cells). The diameter of ex-vivo
generated proerythroblasts is three times greater than that of the corresponding cells generated in vivo. With maturation, the size of ex-vivo generated
erythroblasts becomes similar to that of in-vivo generated cells, although they remain macrocytic (11.6 mm) (the size of RBCs in vivo decreases during
ontogenesis from 20 mm of embryonic erythroblasts to 12.5 mm of fetal erythroblasts and 8 mm of the adult normocytic RBCs) [20]. DXM,
dexamethasone; EPCs, erythroid progenitor cells; EPO, erythropoietin; FSC(s), forward size scatters; HSCs, hematopoietic stem cells; IGF,
insulin-like growth factor; IL, interleukin; MNCs, mononuclear cells; RBC(s), red blood cell(s); SCF, stem cell factor.

addressable with current knowledge. The major obstacles necrosis factor-related apoptosis-inducing ligand, that
in defining an ex-vivo RBC production process, however, inhibit the growth of additional erythroblasts by accel-
cannot be surmounted with current knowledge and erating maturation [51], inducing apoptosis [52] or both
derive from the fact that cultured erythroblasts release (Fig. 4 [52–60]). To reduce the effects of this paracrine
in the supernatant a host of negative regulatory factors loop, erythroblasts must be maintained in culture at
[50], including transforming growth factor beta and tumor densities lower than 106 cells/ml. Therefore, ex-vivo

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Erythroid cells in vitro Migliaccio et al. 265

Table 4 Design considerations for an erythroblasts production process


Parameter

Cell source Cord blood, adult blood, hESCs, iPCs, and others
Number of cells in the transfusion product
RBCs 2.5  1012 per transfusion
Erythroblasts 5  1010 per transfusion
Culture volumes required (l)
RBCs 2500
Erythroblasts 50
Costs (/l) 1000 $
Time required for production (days)
RBCs 30
Erythroblasts 10
The number of RBCs for transfusion is by comparison with the number of RBCs present in a blood unit. The hypothetic number of erythroblasts to be
transfused has been calculated on the basis of the conservative estimate that each erythroblast would undergo at least 5–6 duplications in vivo
(an erythroblast may undergo from four to 64 times before becoming a RBC). The volumes required to produce a transfusion product are calculated on
the basis of the assumption that the cell density within the expansion culture must be kept at 106/ml. Costs are calculated on the basis of the market
price of growth factors considering that the source of EPO may be represented by relatively economic preparations used in the clinic. The time required
for production is calculated on the basis of the speed of expansions in cultures depleted of basoerythroblasts reported in [28]. EPO, erythropoietin;
hESCs, human embryonic stem cells; iPCs, induced pluripotency cells; RBC(s), red blood cell(s).

generation of a unit of blood (2.5  1012 erythroblasts) reported to predict the numbers of erythroblasts that
requires one cubic meter of media (Table 4). The identi- are generated by different cord blood units in cultures
fication of manipulations that increase the density at [63,64] and telomerase expression, low in CD34þ cells,
which erythroblasts can be cultured (i.e. inhibition of increases by several fold with erythroid maturation [65].
the paracrine loop, removal/storage of erythroblasts or
both as they mature) will play a major role in developing Progression of the production process
an ex-vivo RBC production process. Biomarkers that can be used to monitor erythroblasts
expansion during production are represented by anti-
Definition of biomarkers genic, expression profiling or both (Fig. 2). Antigenic
In the case of ex-vivo RBC transfusion products, bio- profiling can be performed using two-color flow cytome-
markers must be developed to predict potency of EPCs try and the levels of CD253a (glycophorin A) and either
from different donors to generate erythroblasts, the pro- CD71 (transferrin receptor type I) [66] or CD36 (throm-
gression of the production process and the potency of bospondin receptor, a receptor for malarial parasites) [38]
ex-vivo generated erythroblasts as transfusion product expression (Fig. 3). Expression profiling may be based on
in vivo. levels of GATA1 expression. In precultures, GATA1/
PU1 and GATA2/GATA1 expression ratios may be used
Potency of erythroid progenitor cells from different donors to as indexes, respectively, of the frequency of EPCs within
generate erythroblasts the progenitor cells and of the numbers of divisions
In contrast to the constant number of erythroblasts that allowed for each EPC. The progeny of HSCs become
are generated in repeated cultures from the same donor, EPCs when expression of GATA1 is higher than that
great variability exists in the number of erythroblasts of PU1, a transcription factor essential for myeloid differ-
generated in cultures from different donors. Therefore, entiation [56]. EPCs retain proliferation activity when
biomarkers must be developed to predict the potency of a expression of GATA2, another member of the GATA
donor to generate erythroblasts ex vivo. Studies are family, is higher than that of GATA1 [67]. On the con-
necessary to establish whether these biomarkers may trary, the level of GATA1 activity may predict if eryth-
be represented by the frequency of EPCs present in cord roblasts in expansion cultures will expand, mature or
blood and adult blood (Table 3), by genetic polymorph- enter apoptosis (Fig. 4).
isms that affect the ability of the EPCs to generate
erythroblasts in culture or both. Candidate polymorphic The potency of the transfusion product in vivo
loci include glucocorticoid receptor [61] and ESR [62] The purpose of the product potency assay is to compare
(Fig. 4) or loci linked to the length of the telomeres potencies of products obtained from different prep-
[63,64]. As both DXM [59] and estradiol [60] target the arations. A potency assay must be based on xenogenetic
levels of GATA1 activity in erythroblasts, different glu- animal models, and human erythroblasts have been
cocorticoid receptor and ESR expression patterns in shown to generate RBCs, although at low frequency,
EPCs from donors may establish a range of alternative in nonobese diabetic/severe combined immunodefi-
synergistic–antagonistic interactions that affects the effi- ciency mice. As the low efficiency may be related to
ciency of the erythroblasts production process (Fig. 4). In the inefficient binding of murine EPO to the human
addition, telomere length of CD34þ cells has been EPO receptor [68], transfusion of ex-vivo generated

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
266 Hematopoiesis

Figure 4 Growth factors and hormones control the fate of the This risk is higher for products that will be administered
erythroid progenitor cells (proliferation, maturation or apopto-
multiple times such as ex-vivo generated RBCs. There-
sis) through discrete changes of GATA1 activity
fore, completely humanized media must be developed
for use in production. Transmission of known and
unknown adventitious agents [71] and neoplastic trans-
formation are also concerns [72–74]. Infection risk can be
addressed following guidelines already developed for
RBC blood products. Although ex-vivo generated eryth-
roblasts stored for 8 years remain chromosomally normal
[75], the safety of ex-vivo generated erythroblasts must
be further evaluated by cytogenetic/genotoxicity assays
in compliance with FDA guidelines (http://www.fda.gov/
Genetic evidence in mice indicates that progression along the erythroid cber/ind/ind.htm).
maturation pathway is controlled by discrete changes in the levels of
GATA1 activity. In fact, both mice carrying the hypomorphic GATA1
mutations [53] and those overexpressing the GATA1 gene [54] die of
anemia. In the case of the hypomorphic mutations, the anemia is induced Conclusion
by increased apoptosis rates at the basophilic erythroblasts stage.
Increased apoptotic rates are due to the fact that in erythroblasts GATA1
Substantial progress has been made in expanding eryth-
controls the expression of the antiapoptotic gene BCLXL [55]. Therefore, roblasts ex vivo, suggesting that ex-vivo generation of RBC
if the GATA1 activity decreases below the thresholds necessary to transfusion products may became a reality in the near
sustain BCLXL expression, erythroblasts will undergo apoptosis. On the
contrary, the anemia in mice overexpressing GATA1 is due to insufficient
future. Much of the research performed in this field is
RBC production because of accelerated maturation. GATA1, by forming directed toward developing hESCs and iPCs that would
complexes with numerous other proteins (FOG1, Lmo2 and many generate erythroblasts customized for the recipient.
others) binds to multiple sites present on the promoter of all the
erythroid-specific genes, activating their expression [56]. Maturation
Development of a clinical grade transfusion product also
occurs when the concentration of GATA1 is sufficiently high to form requires better definition of the identity of the product
these complexes and to saturate all binding sites. Therefore, the rate at (EPC, erythroblasts or RBCs) and the development of
which GATA1 activity increases determines the speed with which
maturation is completed. The anemia observed in mice overexpressing
production processes that permit safe ex-vivo expansion of
GATA1 is due to the fact that in the erythroblasts of these mice GATA1 human erythroblasts in a cost-efficient manner.
concentration increases too rapidly, limiting expansion. These obser-
vations in mice suggests that factors that would either decrease or
increase too rapidly the levels of GATA1 activity will both reduce the Acknowledgements
numbers of erythroblasts generated in vitro by either promoting apop- This study was supported by Ministero per la Ricerca Scientifica, grant
tosis or accelerating maturation. Both types of factors are present in no. RBNE0189JJ_003, and RBNE015P72_003, the National Cancer
expansion cultures. The major positive control on GATA1 activity is Institute, grant no. P01-CA108671 and a New York State STAR grant,
exerted by EPO. EPO stimulates GATA1 biosynthesis by regulating the USA.
transcription rate of the gene and the stability (by inhibiting the caspase
pathway) [57] and phosphorylation state (via the ERK/MAP kinase
pathway) of the protein [58]. Both glucocorticoid receptor and ESR,
instead, negatively regulate GATA1 activity in erythroblasts, glucocorti- References and recommended reading
coid receptor by destabilizing the protein [59], ESR by binding GATA1 Papers of particular interest, published within the annual period of review, have
into transcriptionally inactive complexes [60]. In addition, the paracrine been highlighted as:
production of TGF-b and TRAIL during the culture may also affect  of special interest
GATA1 activity. TGF-b, by increasing GATA1 activity, may accelerate  of outstanding interest
maturation, whereas TRAIL, by activating the caspase 8-dependent Additional references related to this topic can also be found in the Current
degradation of GATA1, may induce apoptosis [52]. Generation of World Literature section in this issue (p. 317).
optimal numbers of erythroblasts in cultures depends on the establish-
ment of a delicate equilibrium among all these factors that ensures that 1 The US Department of Health and Human Services. The 2007 National Blood
the level of GATA1 activity remains within the ranges that allow pro- Collection and Utilization Survey Report. Washington, District of Columbia:
DHHS; 2008.
liferation. EPC, erythroid progenitor cell; EPO, erythropoietin; ESR,
estrogen receptor; MAP, mitogen-activated protein; RBC, red blood 2 Sullivan MT, Cotten R, Read EJ, Wallace EL. Blood collection and transfusion
cell; TGF-b, transforming growth factor beta; TRAIL, tumor necrosis in the United States in 2001. Transfusion 2007; 47:385–394.
factor-related apoptosis-inducing ligand. 3 Nightingale S, Wanamaker V, Silverman B, et al. Use of sentinel sites for daily
monitoring of the US blood supply. Transfusion 2003; 43:364–372.
4 Wallace EL. Monitoring the nation’s blood supply. Transfusion 2003;
43:299–301.
erythroblasts in immunocompromised mouse models
5 Improving blood safety worldwide [editorial]. Lancet 2007; 370:361.
treated with human EPO may represent a more sensitive 6 Bennett C, Silver SM, Djulbegovic B, et al. Venous thromboembolism and
potency assay for these products (Fig. 2). mortality associated with recombinant erythropoietin and darbepoetin admin-
istration for the treatment of cancer-associated anemia. JAMA 2008;
299:914–924.
7 Rizzo JD, Somerfield MR, Hagerty KL, et al. Use of epoetin and darbepoetin in
Safety of the product patients with cancer: 2007 American Society of Clinical Oncology/American
Ongoing clinical trials indicate that immunization of Society of Hematology clinical practice guideline update. J Clin Oncol 2008;
26:132–149.
recipients to xenogeneic products used for cell pro- 8 Zimrin AB, Hess JR. Current issues relating to the transfusion of stored red
duction represents a major risk of cell therapy [69,70]. blood cells. Vox Sang 2009; 96:93–103.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Erythroid cells in vitro Migliaccio et al. 267

9 Reesink HW, Engelfriet CP, Schennach H, et al. International forum: donors 35 Chang AH, Stephan MT, Sadelain M. Stem cell-derived erythroid cells mediate
with a rare pheno (geno) type. Vox Sang 2008; 95:236–253.  long-term systemic protein delivery. Nat Biotechnol 2006; 24:1017–1021.
This paper describes the development of human stromal cell lines.
10 Riley W, Schwei M, McCullough J. The United States’ potential blood donor
pool: estimating the prevalence of donor-exclusion factors on the pool of 36 Bender JG, Unverzagt KL, Walker DE, et al. Identification and comparison of
potential donors. Transfusion 2007; 47:1180–1188. CD34-positive cells and their subpopulations from normal peripheral blood
and bone marrow using multicolor flow cytometry. Blood 1991; 77:2591–
11 Alter HJ, Stramer SL, Dodd RY. Emerging infectious diseases that threaten
2596.
the blood supply. Semin Hematol 2007; 44:32–41.
37 Wong A, Yuen PM, Li K, et al. Cord blood collection before and after placental
12 Flickinger C. In search of red blood cells for alloimmunized patients with sickle
delivery: levels of nucleated cells, haematopoietic progenitor cells, leukocyte
cell disease. Immunohematology 2006; 22:136–142.
subpopulations and macroscopic clots. Bone Marrow Transplant 2001;
13 Crawford SO, Reich NG, An MW, et al. Regional and temporal variation in 27:133–138.
American Red Cross blood donations, 1995 to 2005. Transfusion 2008;
38 Chen L, Gao Z, Zhu J, Rodgers GP. Identification of CD13þCD36þ cells as a
48:1576–1583.
common progenitor for erythroid and myeloid lineages in human bone marrow.
14 Winslow RM. Red cell substitutes. Semin Hematol 2007; 44:51–59; review. Exp Hematol 2007; 35:1047–1055.
15 Garratty G. Modulating the red cell membrane to produce universal/stealth 39 Stellacci E, Di Noia A, Di Baldassarre A, et al. Interaction between the
donor red cells suitable for transfusion. Vox Sang 2008; 94:87–95; review. glucocorticoid and the erythropoietin receptors in human erythroid cells.
16 Neildez-Nguyen TM, Wajcman H, Marden MC, et al. Human erythroid cells Exp Hematol (in press).
produced ex vivo at large scale differentiate into red blood cells in vivo. Nat 40 Chasis JA, Mohandas N. Erythroblastic islands: niches for erythropoiesis.
Biotechnol 2002; 20:467–472.  Blood 2008; 112:470–478.
17 Migliaccio G, Di Pietro R, di Giacomo V, et al. In vitro mass production of A superb review on the biology of the RBC enucleation process.
human erythroid cells from the blood of normal donors and of thalassemic 41 Isern J, Fraser ST, He Z, Baron MH. The fetal liver is a niche for maturation of
patients. Blood Cells Mol Dis 2002; 28:169–180. primitive erythroid cells. Proc Natl Acad Sci U S A 2008; 105:6662–6667.
18 Lu SJ, Feng Q, Park JS, et al. Biological properties and enucleation of 42 Leimberg MJ, Prus E, Konijn AM, Fibach E. Macrophages function as a ferritin
 red blood cells from human embryonic stem cells. Blood 2008; 112:4475– iron source for cultured human erythroid precursors. J Cell Biochem 2008;
4484. 103:1211–1218.
The first observation that erythroid cells derived from hESCs terminally mature
43 Hiroyama T, Miharada K, Sudo K, et al. Establishment of mouse embryonic
in vitro.
 stem cell-derived erythroid progenitor cell lines able to produce functional red
19 Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology. blood cells. PLoS ONE 2008; 3:e1544.
 Cell 2008; 132:631–644. A proof of principle that ex-vivo generated murine RBCs protect from lethal anemia
A superb review on current knowledge on the molecular control of erythropoiesis. in a mouse model of transfusion.
20 Papayannopoulou Th, Abkowitz J, D’Andrea A, Migliaccio AR. Biology of 44 Wozniak RJ, Bresnick EH. Epigenetic control of complex loci during erythro-
eryththropoiesis, erythroid differentiation and maturation. In: Hoffman R, Benz  poiesis. Curr Top Dev Biol 2008; 82:55–83.
EJ, Shattil SJ, et al., editors. Hematology: basic principles and practice, 5th ed. A superb review on the epigenetic regulation of erythropoiesis.
Philadelphia, Pennsylvania, USA: Elsevier; 2009. pp. 276–294.
45 Hochedlinger K, Plath K. Epigenetic reprogramming and induced pluripo-
21 Gursoy A, Dogruk Unal A, Ayturk S, et al. Polycythemia as the first manifesta-  tency. Development 2009; 136:509–523.
tion of Cushing’s disease. J Endocrinol Invest 2006; 29:742–744. An up-to-date review on the development of techniques for generation of induced
22 Dukes PP, Goldwasser E. Inhibition of erythropoiesis by estrogens. Endocri- pluripotency stem cells.
nology 1961; 69:21–29. 46 Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by
23 Golde DW, Bersch N, Cline MJ. Potentiation of erythropoiesis in vitro by defined factors is greatly improved by small-molecule compounds. Nat
dexamethasone. J Clin Invest 1976; 57:57–62. Biotechnol 2008; 26:795–797.

24 Singer JW, Adamson JW. Steroids and hematopoiesis. III. The response of 47 Bhattacharya N. A preliminary report of 123 units of placental umbilical cord
granulocytic and erythroid colony-forming cells to steroids of different classes. whole blood transfusion in HIV-positive patients with anemia and emaciation.
Blood 1976; 48:855–864. Clin Exp Obstet Gynecol 2006; 33:117–121.

25 Bauer A, Tronche F, Wessely O, et al. The glucocorticoid receptor is required 48 Ende M, Ende N. Hematopoietic transplantation by means of fetal (cord)
for stress erythropoiesis. Genes Dev 1999; 13:2996–3002. blood. A new method. Va Med Mon (1918) 1972; 99:276–280.

26 Wessely O, Bauer A, Tran Quang C, et al. A novel way to induce erythroid 49 Migliaccio G, Sanchez M, Masiello F, et al. Dynamic pattern of adhesion
progenitor self renewal: cooperation of c-Kit with the erythropoietin receptor. receptor expression during the maturation of ex-vivo generated human adult
Biol Chem 1999; 380:187–202. and neonatal erythroid cells [abstract]. Blood 2008; 112s:997.

27 Panzenbock B, Bartunek P, Mapara MY, Zenke M. Growth and differentiation 50 Majka M, Janowska-Wieczorek A, Ratajczak J, et al. Numerous growth factors,
of human stem cell factor/erythropoietin-dependent erythroid progenitor cells cytokines, and chemokines are secreted by human CD34(þ) cells, myelo-
in vitro. Blood 1998; 92:3658–3668. blasts, erythroblasts, and megakaryoblasts and regulate normal hematopoi-
esis in an autocrine/paracrine manner. Blood 2001; 97:3075–3085.
28 von Lindern M, Zauner W, Mellitzer G, et al. The glucocorticoid receptor
cooperates with the erythropoietin receptor and c-Kit to enhance and sustain 51 Bohmer RM. IL-3-dependent early erythropoiesis is stimulated by autocrine
proliferation of erythroid progenitors in vitro. Blood 1999; 94:550–559. transforming growth factor beta. Stem Cells 2004; 22:216–224.

29 Giarratana MC, Kobari L, Lapillonne H, et al. Ex vivo generation of fully mature 52 Zamai L, Secchiero P, Pierpaoli S, et al. TNF-related apoptosis-inducing
human red blood cells from hematopoietic stem cells. Nat Biotechnol 2005; ligand (TRAIL) as a negative regulator of normal human erythropoiesis. Blood
23:69–74. 2000; 95:3716–3724.

30 Leberbauer C, Boulme F, Unfried G, et al. Different steroids co-regulate long- 53 Martelli F, Ghinassi B, Panetta B, et al. Variegation of the phenotype induced
term expansion versus terminal differentiation in primary human erythroid by the Gata1low mutation in mice of different genetic backgrounds. Blood
progenitors. Blood 2005; 105:85–94. 2005; 106:4102–4113.

31 Baek EJ, Kim HS, Kim S, et al. In vitro clinical-grade generation of red blood 54 Whyatt D, Lindeboom F, Karis A, et al. An intrinsic but cell-nonautonomous
cells from human umbilical cord blood CD34þ cells. Transfusion 2008; defect in GATA-1-overexpressing mouse erythroid cells. Nature 2000;
48:2235–2245. 406:519–524.

32 Dorn I, Lazar-Karsten P, Boie S, et al. In vitro proliferation and differentiation of 55 Kuo YY, Chang ZF. GATA-1 and Gfi-1B interplay to regulate Bcl-xL tran-
human CD34þ cells from peripheral blood into mature red blood cells with scription. Mol Cell Biol 2007; 27:4261–4272.
two different cell culture systems. Transfusion 2008; 48:1122–1132. 56 Cantor AB, Orkin SH. Transcriptional regulation of erythropoiesis: an affair
33 Miharada K, Hiroyama T, Sudo K, et al. Efficient enucleation of erythroblasts involving multiple partners. Oncogene 2002; 21:3368–3376.
differentiated in vitro from hematopoietic stem and progenitor cells. Nat 57 Ribeil JA, Zermati Y, Vandekerckhove J, et al. Hsp70 regulates erythropoiesis
Biotechnol 2006; 24:1255–1256. by preventing caspase-3-mediated cleavage of GATA-1. Nature 2007;
34 Ledran MH, Krassowska A, Armstrong L, et al. Efficient hematopoietic 445:102–105.
 differentiation of human embryonic stem cells on stromal cells derived from 58 Zhao W, Kitidis C, Fleming MD, et al. Erythropoietin stimulates phosphoryla-
hematopoietic niches. Cell Stem Cell 2008; 3:85–98. tion and activation of GATA-1 via the PI3-kinase/AKT signaling pathway.
This study describes the development of human stromal cell lines. Blood 2006; 107:907–915.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
268 Hematopoiesis

59 Chang TJ, Scher BM, Waxman S, Scher W. Inhibition of mouse GATA-1 67 Grass JA, Boyer ME, Pal S, et al. GATA-1-dependent transcriptional repres-
function by the glucocorticoid receptor: possible mechanism of steroid sion of GATA-2 via disruption of positive autoregulation and domain-wide
inhibition of erythroleukemia cell differentiation. Mol Endocrinol 1993; chromatin remodeling. Proc Natl Acad Sci U S A 2003; 100:8811–8816.
7:528–542. 68 Divoky V, Liu Z, Ryan TM, et al. Mouse model of congenital polycythemia:
60 Blobel GA, Orkin SH. Estrogen-induced apoptosis by inhibition of the homologous replacement of murine gene by mutant human erythropoietin
erythroid transcription factor GATA-1. Mol Cell Biol 1996; 16:1687– receptor gene. Proc Natl Acad Sci USA 2001; 98:986–991.
1694. 69 Selvaggi TA, Walker RE, Fleisher TA. Development of antibodies to fetal calf
61 van Rossum EF, Binder EB, Majer M, et al. Polymorphisms of the glucocorti- serum with arthus-like reactions in human immunodeficiency virus-infected
coid receptor gene and major depression. Biol Psychiatry 2006; 59:681– patients given syngeneic lymphocyte infusions. Blood 1997; 89:776–779.
688. 70 Sakamoto N, Tsuji K, Muul LM, et al. Bovine apolipoprotein B-100 is a
62 Almeida S, Hutz MH. Genetic variation of estrogen metabolism and the risks dominant immunogen in therapeutic cell populations cultured in fetal calf
of cardiovascular disease. Curr Opin Investig Drugs 2007; 8:814–820; serum in mice and humans. Blood 2007; 110:501–508.
review. 71 Turner M. The impact of new-variant Creutzfeldt–Jakob disease on blood
63 Bartolovic K, Balabanov S, Berner B, et al. Clonal heterogeneity in growth transfusion practice. Br J Haematol 1999; 106:842–850.
kinetics of CD34þCD38 human cord blood cells in vitro is correlated with 72 Tolar J, Nauta AJ, Osborn MJ, et al. Sarcoma derived from cultured mesench-
gene expression pattern and telomere length. Stem Cells 2005; 23:946– ymal stem cells. Stem Cells 2007; 25:371–379.
957.
73 Rubio D, Garcia-Castro J, Martin MC, et al. Spontaneous human adult stem
64 Schuller CE, Jankowski K, Mackenzie KL. Telomere length of cord blood- cell transformation. Cancer Res 2005; 65:3035–3039.
derived CD34(þ) progenitors predicts erythroid proliferative potential.
74 Bernardo ME, Zaffaroni N, Novara F, et al. Human bone marrow derived
Leukemia 2007; 21:983–991.
mesenchymal stem cells do not undergo transformation after long-term in vitro
65 Prade-Houdellier N, Frebet E, Demur C, et al. Human telomerase is regulated culture and do not exhibit telomere maintenance mechanisms. Cancer Res
by erythropoietin and transforming growth factor-beta in human erythroid 2007; 67:9142–9149.
progenitor cells. Leukemia 2007; 21:2304–2310. 75 Sanchez M, Leblanc A, Mancini A, et al. Human erythroblast generated in vivo
66 Kato J, Kobune M, Ohkubo S, et al. Iron/IRP-1-dependent regulation of mRNA remain functional with a normal karyotype 8 years after cryopreservation:
expression for transferrin receptor, DMT1 and ferritin during human erythroid implications for ex vivo generated erythroid transfusion product [abstract].
differentiation. Exp Hematol 2007; 35:879–887. Blood 2008; 112s:2303.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like