Sie sind auf Seite 1von 21

Journal of Stem Cells ISSN: 1556-8539

Volume 11, Number 3 2016 Nova Science Publishers, Inc.

Stage-Specific Regulation of Erythropoiesis and Its


Implications in Ex-Vivo RBCs Generation

Vimal Kishor Singh1,, Abhishek Saini1, Abstract


Manisha Kalsan1, Neeraj Kumar1, and
Ramesh Chandra2 Ex vivo erythropoiesis methods are being developed for
1 more than a decade now, and all the distinct types of stem
Stem Cell Research Laboratory, Department of cells (such as CD34+ HSCs, ESCs, IPSCs, and extensively
Biotechnology, Delhi Technological University, proliferating erythropoietic progenitor cells) are defined to
Delhi, India bear the potential for large scale RBC production shortly.
2
Dr. B. R. Ambedkar Center for Biomedical Research, The various regulating factors at different levels of RBCs
University of Delhi, Delhi, India production are being explored. Since most of the ex-vivo
erythropoiesis protocols mimic the dogma followed by
hematopoietic stem cells in vivo to give rise to mature
RBCs which essentially deals with the intermediate stages
of erythropoiesis such as burst forming unit-erythroid
(BFU-E) and committed erythroid colony forming unit-
erythroid (CFU-E). In vivo generation of erythroid
progenitors (BFU-E/CFU-E) is essentially controlled by
several factors including glucocorticoids, inflammation, and
stress. Furthermore, regular production of functionally
mature /transfusable units of RBCs is possible only through
the coordinated regulation of terminal proliferation and
differentiation of erythroid progenitors by external signals,
such as erythropoietin, SCF, IL-3 and interaction to
extracellular matrix protein(s) in a 3D culture system. We
discuss these complex intracellular networks of coordinated
factors and try to understand their molecular mechanism
through gene regulation by transcription factors, and
miRNAs that might be helpful in developing the optimal
RBCs production protocols for commercial production.

Abbreviations
EPO Erythropoietin
SCF Stem Cell Factor
IL3 Interleukin-3
TPO Thrombopoietin
Flt3 Fms related Tyrosine Kinase 3
IGF1 Insulin like Growth Factor 1
Correspondence: Dr. Vimal Kishor Singh: O/I, Stem cell Sox2 SRY (Sex determining region Y)-
Research Laboratory, Department of Biotechnology, box-2
Delhi Technological University, Shahbad Daulatpur, miRNAs micro RNA(Ribose Nucleic Acid)
Bawana road, Delhi-110042. shRNA short hairpin RNA
E-mails: vim_kissor@yahoo.co.in;
vimalkishorsingh@gmail.com, vimalksingh@dce.ac.in
3D Three Dimensional
150 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

WHO World Health Organization circulatory system and developed ways to study the
RBC Red Blood Cells cross circulatory effects in animals. Later on Dr.
HSCs Hematopoietic Stem Cells James Blundel (1818) reported first successful blood
ESCs Embryonic Stem Cells transfusion among humans. In 1901, Karl Landsteiner
hESCs human Embryonic Stem Cells discovered the blood group antigens and also
iPSCs induced Pluripotent Stem Cells demonstrated the adverse effects associated with the
UCB Umbilical Cord Blood transfusion of incompatible blood groups in human
PB Peripheral Blood recipients leading to severe immunological disorders
BM Bone Marrow causing death. This landmark study (Nobel Prize
CD34 Cluster of Differentiation 34
1930) laid the foundation for the identification of
CD235a Cluster of Differentiation 235a
various blood type antigens and paved the way for
(Glycophorin A)
safe and fruitful clinical blood transfusions. Later
CFU-E Colony Forming Unit Erythroid
studies demonstrated a number of different blood cell
BFU-E Burst Forming Unit Erythroid
MCSF Macrophage Colony Stimulating surface antigens playing a crucial role in determining
Factor the compatibility of donor-derived blood in clinics.
GCSF Granulocyte Colony Stimulating In 19401950s, a number of inventions made it
Factor possible to store the donor-derived human blood
Stat5 Signal Transducer and Activator of under appropriate conditions, and a vast network of
Transcription 5 blood banking system flourished during these years.
GATA 1 Globlin Transcription Factor 1 In modern medical modalities, blood bank system
GATA 2 Globlin Transcription Factor 2 remains axial to support all kinds of blood transfusion
SCL/Tal1 Stem Cell Leukemia/ T-cell Acute needs (Alter and Klein, 2008). There are about 108
Lymphocytic Leukemia 1 million blood units collected throughout the globe
LDB1 LIM Domain Binding 1 every year (www.who.int/worldblooddonorday/en/)
LMO2 LIM Domain Only 2 (Department of Health and Human Services, 2010,
Klf1 Kruppel Like Factor1 2013; World Health Organization, 2011). However,
Gfi-1b Growth Factor Independent 1b the accessibility to safe and adequate blood supplies is
transcription Repressor limited in developing countries as compared to the
MYB Myeloblastosis developed countries. Developed countries rather
ALK Anaplastic Lymphoma Kinase sparsely populated (15% of the global population) but
contribute to more than 50% of the total global blood
collection per annum. This is the reason that problem
Introduction of shortage of transfusable blood units is not so severe
in the economically developed countries, and the
In the17th century, Dr. Harvey established a blood collection seems to be sufficient for the time
breakthrough by reporting his findings regarding being. According to the WHO reports, there are
blood circulation system that eventually led to the approximately 8000 blood centers scattered in 159
blood transfusion experiments. Initial blood high-income countries which score >30,000 annual
transfusion experiments were done on animals that blood donations on an average per blood center
laid the foundation for human transfusion (Department of Health and Human Services, 2010,
experiments. In 1665, Dr. Jean-Baptiste Denys 2013). On the other hand, only 3700 blood collections
demonstrated first successful human transfusion from per center are reported in developing countries which
a sheep in 15 years old boy. Unfortunately, this have to support ~ 80% of the world population
experimentation of animal blood transfusion in residing in these countries. The WHO reports states
humans resulted in the death of most of the recipients that there are only ten donations per 1000 people in
as reported by him in successive studies. Another the 82 low income and middle-income countries
milestone was achieved by Dr. Richard Lower, who (World Health Organization, 2011) (Figure 1).
showed the effects of changes in blood volume in the
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 151

Figure 1. Graphical presentation showing a total number of blood donations, blood donations in developed vs. developing
countries. The population of developed countries is about 18% who have access to approximately 50% of the total donated
blood. Whereas, the developing countries where more than 80% of the global population have access to the remaining 50%
of the donated blood.

What makes the situation much horrible is the hemoglobinopathies, polytransfusion patients, and
inefficient screening facilities available in most of the polyimmunization may put more constraints on the
undeveloped countries. A significant number of donor derived blood supply system across the globe
countries (39 countries) are reported to lack efficient (Table 1) (World Health Organization, 2011).
screening facilities for most severe transfusion- Although, there are reports showing methods to
transmissible infections (TTIs) viz. HIV, hepatitis B, develop universal blood group by using antigen
hepatitis C, and syphilis (Table 1) (Department of masking and/or enzymatic cleavage techniques but
Health and Human Services, 2013). Further, only 106 these methods are yet to be elaborated in a detailed
countries have laid national guidelines for appropriate way (Bagnis et al., 2011).
blood transfusions, and only 13% of the low-income Another important issue is the association of
countries are equipped with the national various types of adverse reactions with the donor
haemovigilance system to ensure the safety of the derived blood supplies. Despite a number of testing,
transfusion process. It is also interesting to be noticed preclinical precautionary measures taken to avoid any
that most developed countries are having a rapidly infectious agent [e.g., HIV-1, HIV-2, HTLV-1,
growing proportion of elderly people (>60 years old). HTLV-2, Hepatitis B, Hepatitis C, Syphilis (T.
Moreover, it would be difficult to support the need of pallidum), Chagas disease (T. cruzi), and West Nile
burgeoning demand for blood transfusions for Virus, Cytomegalovirus (CMV)], blood transfusions
surgical treatments by the year of 2050 (U.S. Census are prone to several complicated situations (Alter and
Bureau, 2004; Ali et al., 2010). The situation would Klein, 2008; World Health Organization, 2011).
be more difficult if one has to define the blood group These complications cause delayed cure rate with
compatibility for more than 30 blood group system prolonged hospitalization periods and may raise the
(308 recognized antigens) including ABO & Rh overall cost of treatment also (Department of Health
antigens (Daniels et al., 2009). Also, demand for and Human Services, 2010).
rare phenotypic blood, patients with various
152 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

Table 1. Risks associated with the blood transfusion

S.No. Agent Risk Factor Remarks


HIV 1 in 4 million
Hepatitis B attacks liver 1 in 1.2 million
1. Blood borne infections
Hepatitis C 1 in 3 million
West Nile Virus 1 in 1 million
Swelling
2. Allergies Can be treated with antihistamines
Itching
Maybe due to febrile transfusion
Fever
Body temperature reaction
3.
fluctuation Due to temperature difference of blood
Hypothermia
being transfused
The white cells in the transfused
4. Graft versus host disease Use of irradiated blood prevents it
blood attack patients tissue
Acute immune hemolytic Patients immune system destroys
5. Very rare but serious due to mismatch.
reaction the transfused blood cells

Figure 2. Timeline Schematic representation of the major events related to ex-vivo erythropoiesis generation by different
research groups highlighting there break-troughs.

For the last few years, various non-donor derived carriers and have limited functional application
sources have been proposed by different research (Winslow, 2006; HenkelHonke and Oleck, 2007;
groups. However, the majority of artificial blood Natanson et al., 2008; Silverman and Weiskopf, 2009;
molecules (hemoglobin-based oxygen carriers or Castro and Briceno, 2010). One of the most promising
perfluorocarbon solutions) are inefficient oxygen approaches is ex vivo expansion of erythrocytes from
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 153

stem cells. Initially, Nakamura and his colleague nucleated erythroid precursors in vivo through their
demonstrated survival of lethally irradiated mice by interaction with internal factors regulating their
ex vivo expanded RBCs which were derived from maturation. This fact is well supported by the results
immobilized ESCs cell line (Hiroyama et al., 2008). from studies reported both in human and animal
Following the Nakamuras findings, many people models (Ende and Ende, 1972; Neildez-Nguyen et al.,
have developed different protocols to derive 2002). The significance of the maturing erythrocytic
functionally mature erythrocytes from various types precursor is also supported by several reports showing
of stem cells viz. CD34+ HSCs, Embryonic stem the occasional clinical use of compatible Cord Blood
cells, and IPSCs. The proof-of-concept study (40-80 ml/47 1010RBCs and 28 109 erythroblast
demonstrated by Giarratana et al., shown first-in- cells) for transfusion in emergent conditions (Ende
human blood transfusion of ex-vivo generated RBCs and Ende, 1972). It is evident from these reports that
(Giarratana et al., 2011). These studies indicated that ex vivo RBCs expansion can support the different
expansion of erythrocytes from stem cells could clinical short-comes that exist because of supply
become a powerful tool to solve the problems of constraints and many rare phenotypes related blood-
blood supply shortages and other associated issues. based disorder. However various technological
However, there are several important matters barriers exist, and a regular large scale production
which need to be resolved to ensure the expected would only be possible if one can find the most
yield of functionally mature RBCs through these suitable way to produce clinical transfusable units at a
protocols suitable for clinical transfusions. reasonable cost. To enhance the present scale of
Fortunately, there are reports showing possibilities of RBCs production more cost-effective and timely
exponential expansion of erythrocyte precursors by automated ex vivo culture approaches are required.
using similar techniques. Neildez-Nguyen et al. This is possible if we have an in-depth knowledge of
demonstrated significant yield (up to 0.5 transfusion the various regulatory elements and their mode of
unit) from UCB derived CD34+ cells. An action playing an axial role in almost all of these
approximately 2 million fold expansions of methods demonstrated so far.
enucleated erythrocytes was reported in their studies Here we present the various essential regulator
showing functional similarities between native RBCs elements playing an important role during in vivo
and cultured RBCs (Neildez-Nguyen et al., 2002). In erythropoiesis, and their optimal use may aid to
similar studies, UCB derived CD34+ are reported to develop more efficient large-scale expansion
give rise to 10 transfusion unit by a technically protocols.
complex protocol (Fujimi et al., 2008) (Figure 2).
Despite their ill-suitedness for the large scale
commercial production, these methods precede the Erythropoiesis in a Glance
potential use of stem cell-derived methods in near
future. Apart from overall yield arising from these Mammalian definitive erythropoiesis is initiated
protocols, the enucleation efficiency of maturing in the fetal liver/yolk sac and later on it remains
RBCs has been reported to be significantly low which limited to the adult bone marrow throughout the life.
is a major indicator of their functional maturation. This highly regulated process involves hematopoietic
However, it was demonstrated RBCs precursors, progenitor cells and their subsequent differentiation
cellular intermediates/nucleated erythroid precursors into committed erythroid progenitors. After that, these
generated during ex vivo expansion may also be of use progenitor cells proliferate further and give rise to
if transfused. It is proposed that these maturing functional erythrocytes. Hematopoietic stem cells
nucleated cells along with enucleated erythrocytes can (HSCs) proliferate in the medulla of the adult bone
function like filler supporting the cellular content marrow with high self-renew capabilities and
during the transfusion and may give rise to differentiate into myeloid and lymphoid progenitor
functionally matured cell by proliferating into 464 cells. Lymphoid progenitor cells undergo
enucleated RBCs. This is reflected by studies showing differentiation and give rise to lymphocytes and
the functional maturation of cellular intermediates/ plasma. A myeloid progenitor cell is a multipotent
154 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

progenitor and further down the line differentiates phase, the burst forming unit erythroid (BFU-E) (slow
into bipotential progenitors which are restricted to proliferating) are the earliest committed progenitors
erythroid/megakaryocyte and granulocyte / macro- that can be identified ex vivo. Early BFU-E cells get
phage pathways (Akashi et al., 2000). The hierarchy matured and then differentiated into rapidly dividing
of erythropoiesis may be defined in three distinct stage of colony forming unit-erythroid (CFU-E)
stages (Figure 3). First, the commitment of HSCs into (Elliott et al., 2008). In the next few days, these CFU-
erythropoietic lineages which result to the E progenitors divide further (3-5times) and
development of erythroid-committed blast cells from differentiate into later stages. This includes various
multipotent hematopoietic progenitors; second, significant changes such as reduced cell size,
Division and differentiation of these morphologically chromatin condensation, and synthesis of hemoglobin
distinguishable erythroid progenitor cells; and third, that leads to their enucleation and expulsion of other
Terminal differentiation that includes a change in cell organelles (Fawcett et al., 1997).
morphology viz. removal of nucleus or enucleation to
produce mature RBCs. During erythroid commitment

Figure 3. In vivo erythropoiesis: This schematic diagram represents the three major phases of in-vivo erythropoiesis a)
Phase1- Hematopoietic stem cells differentiate into erythroid precursor BFU-E, b) Phase2- Expansion and differentiation of
BFU-E into erythroid progenitors CFU-E in the presence of IL3, EPO, c) Phase3- Maturation of erythrocytes through
enucleation the reticulocytes are transformed into mature enucleated erythrocytes by the expulsion of the nucleus.

Human RBCs turnover is approximately 1% per CFU-E cells are required since they are not capable of
day which gets substantially increased several times giving rise to such an extreme RBCs population even
under stress conditions such as trauma or hemolysis. in the presence of high concentration of Epo. BFU-E
This stress-induced short term enhanced cells which have receptors for Epo, Stem Cell Factor
erythropoiesis is majorly regulated by Erythropoietin (SCF), IL-3, insulin-like growth factor 1 (IGF-1),
(EPO). EPO is secreted by kidney cells under hypoxic glucocorticoids (GCs), and IL-6, get proliferated and
conditions and stimulates the terminal proliferation differentiated into a number of CFU-E cells to meet
and differentiation of CFU-E progenitors (Molineux the demand. However, their origin in fetal/bone
et al., 2009). Under chronic stress conditions, more marrow has not been defined. The regulatory
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 155

mechanism for the division of BFU-E cells, another commitment, (2) expansion, and (3) maturation.
cytokine interaction essential to control their self- These protocols are well summarized by Singh et al.,
renewal and differentiation into mature CFU-E (2014), and various cytokines/growth factors (SCF
progenitors is not properly understood. There are and EPO are most common) are demonstrated to
many intracellular signal transduction molecules generate variable yields of immature progenitors or
including protein-ligand to the above-mentioned mature RBCs (Singh et al., 2014). Similarly, studies
cytokines/ growth factor receptors, which may elicit are done showing variable effects of animal/human
activation of various downstream transcription derived feeder co-culture system on the yield and
factors, DNA-binding molecules and chromatin maturation level of cultured RBCs. Most of the
modifiers to control cellular division and protocols demonstrated so far have followed the
differentiation. Besides, multiple noncoding dogma in which stem cells are converted into
regulatory RNAs, (microRNAs); are essential for committed erythroid lineages through hematopoietic
these function. cell progenitors states but recently trans-
Thus, the overall in vivo RBC generation process differentiation procedures are reported showing direct
is conceived to be a well-regulated system that relies differentiation of human fibroblast cells into
on cytokines, transcription factors, and cofactors, erythroblasts (Szabo et al., 2010).
post-translational modifications, and miRNAs at Although promising all these processes reported
specific stages. A detailed understanding of these so far inherit almost similar problems including
regulating elements could be helpful in designing inefficient enucleation of erythrocytes, less overall
optimal protocols for large scale RBCs production by yield, lack of expression of an adult form of Hb
manipulating ex vivo erythropoietic culture states predominantly, use of animal derived products such
through these molecules. as serum and feeder cell co-culture. Apart from them,
there is clinical transfusion related problems also such
as difficulty in matching appropriate blood type and
Ex Vivo Expansion of RBCs unavailability of sufficient number of RBCs for
transfusion.
The methods for developing terminally
differentiated, fully functional RBCs in transfusable
amount have evolved enough during last decade Molecular Regulators
(Migliaccio et al., 2012). There have been significant for Erythropoiesis
studies defining various regulatory elements to access
biological control over the erythropoietic process. As Many groups tried to categorize various key
discussed in the next section, studies to define in vivo regulators of ex-vivo RBC expansion in a certain way.
hematopoietic development process have shown that There are several factors which play pivotal roles
the generation of erythroblast is regulated by various including cytokines (e.g., EPO, SCF, and IL3),
growth factors (Lodish et al., 2010). These reports transcription factors, co-factors and non-coding
helped researchers to explore the enormous potential regulatory RNAs (Table 2, Figure 4). All these factors
inherited by all stem cells types including (i) CD34+ affect different stages of ex-vivo erythropoiesis. Some
HSPCs, (II) Embryonic stem cells/induced pluripotent of them may regulate the initial growth and
stem cells, and (III) immortalized erythroid differentiation of most primitive HSCs (e.g., SCF, IL-
precursors. 3) which are the source in almost all protocols. The
All these cell types are shown to get addition of other molecules such as steroids,
differentiated into erythroid lineages in different erythropoietin may be essential for the maintenance of
protocols which all share almost similar growth growth and maturation of erythroid progenitors/
regulators (Figure 4) (Table 2). Despite significant precursors and mature RBCs respectively. These
differences in their methodologies used, time of molecular regulators have a distinct role in these
culture and yield, all of these methods are categorized ex vivo expansion approaches. The overall
in three basic/ major steps as follows: (1) erythropoiesis can be categorized under normal
156 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

regulatory conditions and the regulation of same Whereas, stress conditions is defined in the second
under stress condition. Like the first category, the category which involves a somewhat different
normal erythropoietic condition is regulated by a mechanism to maintain the sudden need of
number of different regulatory elements as discussed hyperproliferative erythropoiesis and has been
below i.e., growth factors, proteins and miRNAs. discussed in the later part.

Figure 4. Insight to erythropoiesis with respect to ex-vivo erythropoiesis: This schematic diagram represents the site of
erythropoiesis i.e., bone marrow in adult human where differentiation of HSCs into erythroid progenitors takes place in an
organized manner under the regulation of various sets of cytokines, transcription factors, and miRNAs which finally matures
into erythrocytes through enucleation of reticulocytes and gets into the circulation. For ex-vivo erythropoiesis, the essentially
required are the source of HSCs which could be UCB, iPSCs, PB which has to be cultured in the presence of growth factors
(blue), chemical inducers (Green) in a span of 18 -21 days to get mature erythrocytes.
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 157

Table 2. Factors that regulate ex-vivo erythropoiesis

S.
Factors Role/Effect in Ex-Vivo Erythropoiesis Receptor References
No.
1 Animal/ Human-Derived Growth Factors/Cytokines
Differentiation and proliferation of
a) EPO EPOR Gregory et al., 1978
erythroid
Differentiation and proliferation of Korpelainen et al.,
b) IL3 CD123/IL3RA,CD131/IL3RB
myeloid progenitor cells 1996
c) GM-CSF White blood cells growth factor CD116 Rosas et al., 2007
Inducer of HSCs mobilization from the
d) G-CSF CD114 Basu et al., 2002
bone marrow into the bloodstream
stimulate erythroid colony forming units in
e) IGF-1 IGF1 Receptor Brox et al.,1996
the mouse
f) SCF Regulates HSCs in the bone marrow CD117 Linden et al., 2004
2 Synthetic Molecules
a) Dex Anti inflammation Leberbauer et al., 2005
b) Poloxamer 88 Induces enucleation of RBCs Baek et al., 2009
Cholestrol
c) Induces proliferation rate Leberbauer et al., 2005
rich Lipid
d) Hydrocortisone Enhance the proliferation of EPCs Timmins et al., 2011
3 mi-RNAs
Differentiation towards the
a) miR-150 megakaryocytic lineage at the expense of MYB Lu et al., 2008
erythroid lineage
Downregulation of miR-221 and miR222
miR-221
b) is required for terminal erythroid KIT Felli et al., 2005
and miR-222
progenitor proliferation
Downregulation of miR-223 is required for
c) miR-223 ALK4 Felli et al., 2009
the terminal differentiation
Downregulation of miR-24 is required for
d) miR-24 LMO-2 Wang et al., 2008
terminal erythroid differentiation
4 Transcription Factors
Globin, erythroid specific
regulates p21 gene expression during
a) GATA1 membrane proteins, Papetti et al., 2010
erythroid differentiation
GATA-1, GATA-2
the cellular proliferation of non-committed Orkin et al., 1992 &
b) GATA2 GATA-1 and GATA-2 gene
and committed erythroid progenitors 2000
required for specification of HSCs and
Ravet et al., 2004; Hall
c) SCL/TAL1 maturation of the erythroid and Glycophorin A, p21 gene
et al., 2005
megakaryocytic lineages
d) LDB1/LMO2 Ldb1 complexes regulate Klf1 Love et al., 2014
Acts as global regulator of erythroid
e) Klf1 Globin Micheal et al., 2010
production and integrity
f) Gfi1B represses p21, SOCS1, 3 and its own genes p21, SOCS1-3, BclxL Kuo et al., 2007

Category-I: Erythropoiesis under Normal is considered as a regulator of erythroid lineage alone


Conditions is insufficient for large scale amplification of HSCs
(Munugalavadla et al., 2005; Arcasoy et al., 2005).
a. Role of Growth Factors Therefore, EPO-supplemented with different
Different research groups working on RBC cytokines viz. SCF, IL-3, Flt-3 & TPO seem to be
production reported that Erythropoietin (EPO) which most promising (Kawada et al., 1999; Jiang et al.,
2006; Drouet et al., 1999).
158 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

EPO response to EPO due to fewer receptors. Further,


Erythropoietin (EPO) a glycoprotein hormone down the erythroid differentiation line, EPOR per cell
that is produced by peritubular capillary lining cells in expression is gradually reduced, and according to the
the kidney and liver. It acts on its target cells present studies the reticulocyte and erythrocyte are devoid of
in the bone marrow to regulate the proliferation, EPO receptors (James 2003). EPO binds EPOR to
differentiation, and maturation of red blood cells activate dimerized EPOR and trigger a cascade of
(Gregory et al., 1978). It is an approximately 30- events viz. change in EPORs conformation and trans-
34kDa glycoprotein. The function of EPO depends on phosphorylation of JAK2 resulting in phosphorylation
the conditions such as availability of oxygen, iron and of multiple tyrosine residues in the cytoplasmic tail of
cofactors, sufficient number of responsive progenitor EPOR. These phosphorylated tyrosine molecules are
cells, and optimal microenvironment. It does not responsible for activation of downstream signaling
specify the cell fate directly into the erythroid lineage pathways such as STAT5, PI3-kinase/AKT, and Ras/
but supports the growth, differentiation and survival MAPK. All these pathways are essential for erythroid
of the erythroid committed progenitor cells (Koury et expansion and maturation. The primitive erythroid
al., 1990). EPO receptor (EPOR) is primarily progenitor, i.e., BFU-E has a low sensitivity for EPO.
expressed on erythroid cells. Its expression is highest Therefore, additional growth factors (SCF, IL-3)
from CFU-E to pro-erythroblast of erythroid cell come in the play for survival and proliferation of cells
development; whereas BFU-E shows a weaker (Lindern et al., 2004) (Table 2, Figure 5).

Figure 5. Regulation of erythropoiesis by growth factors: Differentiation of common myeloid progenitor CFU-GEMM into
erythroid lineage is governed by IL3, GM-CSF, and SCF. First cell in erythroid lineage is burst forming unit- erythroid
(BFU-E) which is EPO responsive and thereafter EPO regulates the maturation of erythroblasts into mature erythrocytes.

BMP4 of BMP4/Smad-4 mediated signaling pathways.


(Singbrant et al., 2010; Singbrant et al., 2006).
Another important factor having an essential role Furthermore, the BMP4 expression is induced by
in stress erythropoiesis is Bone morphogenetic protein hypoxia in spleen stromal cells (Wu et al. 2010;
4 (BMP4). Studies in mice models expressing a Millot et al., 2010). BFU-E cells need priming with
dominant negative Smad5 mutant from that inhibits Sonic Hedgehog to be responsive to BMP4 which is a
BMP4 signaling have shown neonatal anemic morphogen synthesized and secreted spleen cells
characteristics. That lasts for two weeks after birth (Perry et al., 2009).
and then resolved (Heg et al., 2007; Lenox et al., On their activation with Sonic Hedgehog the
2005; Subramanian et al., 2008). transcription factor Smad5 gets activated through
This suggests that BMP4 signaling through BMP-4. BMP-4 also induces activities of Scl and
Smad5 is essential for stress-induced erythropoiesis Gata2 as well and thus enhances the probability of
and adult or fetal liver hematopoiesis is independent BFU-E self-renewal. (Lugus et al., 2007; Fuchs et al.,
2002; Harandi et al., 2010).
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 159

Stem Cell Factor Now the major question arises how to deploy
SCF is the ligand for the c-kit cell-surface these growth factors in ex-vivo erythropoiesis.
receptor or the tyrosine kinase receptor. In anemic Various groups reported different concentrations and
mice, hemoglobin concentration increases to a modest combinations of cytokines. Neildez-Nguyen et al.,
degree upon addition of exogenous EPO due of (2002) reported FLT-3, SCF, TPO, EPO, IGF-1
defects in either SCF or c-kit (Pharr et al., 2000). SCF (Neildez-Nguyen et al., 2002) in similar studies.
works in coordination with other cytokines like GM- Later, Giarratana et al., (2005) shown that only SCF,
CSF (Table 2, Figure 5) whose function is to EPO, and hydrocortisone were sufficient. Leberbauer
stimulate the growth and proliferation of granulocyte et al., (2005) also reported a set of EPO, SCF, IGF-1
and macrophage progenitor cells. It also influences Dex and lipid mix (Leberbauer et al., 2005). Baek et
the differentiation, induce maturation in HSCs and al. (2008-09) have shown similar effects by using
later stimulate the functional activity of mature HSCs SCF, IL-3, EPO, TPO and Flt-3 in their experiments
(Inaba et al., 1992; Metcalf et al., 1995; Rosas et al., (Baek et al., 2009). Other groups used a different set
2007). Another important molecule, IL-3 stimulates of growth factors and certain hormones, steroids, and
the generation and functional activities of various chemicals which point out that cytokines alone are not
blood cell types (Korpelainen et al., 1996). MCSF enough, and they need helping hand. A possible
looks after the proliferation, differentiation, and explanation for that may be the effect of cytokine is
survival of macrophages, monocytes and bone dependent on different cell stages. Therefore, it
marrow progenitor cells (Stanley et al., 1997). G-CSF becomes necessary to choose the most appropriate set
is another glycoprotein playing major role bone of cytokines for culturing the clinical grade
marrow stimulation and production of granulocytes. It erythrocytes.
interaction is also important for their gradual release
into the bloodstream (Basu et al., 2002).

Figure 6. Regulation of erythropoiesis by different proteins and miRNAs. The intarcellular fators such as transcriptions
factors and miRNAs rgulate the formation of RBCs. GATA2 is activity is downregulated by GATA1 to regulate in
erythrocytic determination and terminal differentiation. Other proteins such as SCL/TAL1, Lmo-2, and Ldb1 aid this
regulation. miRNAs come into the action starting from the initial stage where miR-150 down-regulates expression of Myb
gene to inhibit erythroid lineage differentiation. During terminal maturation these miRNAs enhances terminal differentiation
by down regulating the KIT gene that regulates proliferation of erythroid progenitors.
160 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

b. Regulation by Proteins whereas it is associated with an ETS (E26


During erythropoiesis binding of most important transformation-specific) binding site in mega-
growth factor elicit pattern of intracellular proteins. karyocytic specific genes (Orkin et al., 2000; Orkin et
For example, several transcription factors are al., 1992; Papetti et al., 2010). The GATA family of
activated like, Stat5 and EPO-regulated tran- transcription factors consists of six transcription
scriptional regulators present in erythroid cells factors, GATA- 1 to -6 that can bind the consensus
interact with few lineages restricted transcriptional sequence 5 A/T GATA A/G 3. These proteins
regulators such as GATA-1 (Globin Transcription contain two conserved zinc finger motifs (Cys-X2-
Factor 1), SCL/Tal1 (Stem cell leukemia/T-Cell Cys-X17-Cys-X2-Cys) peculiar to the GATA family.
Acute Lymphocytic Leukemia 1), LDB1 (LIM The carboxyl terminal zinc finger binds to DNA,
Domain Binding 1), LMO2 (LIM Domain Only 2), whereas the amino-terminal zinc finger stabilizes the
Klf1(Kruppel-Like Factor 1 ), and Gfi-1b (Growth DNA/GATA interaction. Outside of the zinc finger
Factor-Independent 1B Transcription Repressor), regions, the conservation between GATA factors is
responsible for the production of mRNAs that are low, but each factor is conserved between
essential for erythropoiesis (Table 2, Figure 6). species. GATA-1 and GATA- 2 reported to regulate
GATA1 belongs to the GATA family of transcription in erythrocytic determination and terminal
factors which regulates the switch of fetal hemoglobin differentiation.
to adult hemoglobin in erythroid development. While This is supported by motif analysis reports
the stem cell leukemia (SCL) gene, which is also showing activation of genes enriched consensus
known as Tal-1 encodes a critical regulator of both sequences for SCL/Tal1 at GATA1-bound sites.
hemopoiesis and vasculogenesis i.e., a basic helix These studies suggested correlation between GATA1
loophelix (bHLH) transcription factor and, therefore, activation and SCL/Tal1 activities. (Fujiwara et al.,
positively regulates erythroid differentiation. Next 2009; Yu et al., 2009; Cheng et al., 2009; Kassouf
LDB1 acts with LMO2 by binding to the LIM domain et al., 2010; Tripic et al., 2009; Ravet et al., 2004;
of a transcription factor in order to regulate red blood Hall et al., 2003). In erythroid cells, SCL/Tal1
cell development by maintaining erythroid precursors interacts with basic helix loop helix protein E2A
in their immature state. LMO2 is a cysteine-rich, two (bHLH protein E2A) and makes an activation
LIM-domain protein that has a central and crucial role complex by interacting with LIM domain of LMO2
in hematopoietic development along with TAL1/SCL and Ldb1. Along with GATA1, this complex binds to
and is highly conserved. Klf1 is a hematopoietic- the GATA1/E-box domains that are spaced ~9 to 11
specific transcription factor and induces high nucleotides apart (Elliot et al., 2008). Components
expression of adult beta-globin and other erythroid other than GATA1 in the activation complex are also
genes. Gfi-1b is an essential proto-oncogenic necessary for RBC development. This is indicated by
transcriptional regulator which controls the expression studies in Lmo2, GATA1, and SCL/Tal1 knockout
of genes involved in development and differentiation mice where all such mice are anemic and die due to
of erythrocytes and megakaryocytes by forming lack of primitive erythropoiesis in fetal development
complexes with transcriptional regulatory proteins (Cantor et al., 2002). TAL-1 can assemble in a
including GATA-1, histone deacetylases, and runt- pentameric complex (Wadman et al., 1997)
related transcription factor-1. containing the ubiquitously expressed.
The expression pattern of specific genes is E proteins (E2A, HEB, E2-2), LMO-2, Ldb, and
regulated in a precise time frame at the transcriptional GATA-1 in erythroid-committed cells. This complex
level during the differentiation of erythroid lineage. can lead to activation of erythroid-specific terminal
Functional analysis of erythroid and megakaryocytic genes expression. GATA-2 variant of this TAL-1
specific genes has shown the importance of a complex is responsible for activation of c-Kit
sequence, 5 A/T GATA A/G 3, now called the transcription in CD34+ hematopoietic progenitors
GATA motif, in the lineage-specific expression of (Lecuyer et al., 2002; Love et al., 2014; Michael et
these genes. This sequence is associated with a GT or al., 2010; Kuo et al., 2007). Therefore, to mimic
CACC-like sequence in erythroid-specific genes,
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 161

erythropoiesis ex-vivo a better understanding at process is governed by several important factors


molecular level becomes inevitable. which tightly regulates survival, proliferation and
terminal differentiation of erythroid progenitors such
c. Regulation by miRNAs as CFU-E and produce more number of erythroblast
In the last few years, a new class of small, cells. Since limited proliferative potential (3-5cell
endogenous, non-coding RNAs called microRNAs divisions) of early EPO-responding CFU-E even in
(miRNAs) is reported. miRNAs are important the presence of high level of EPO due to stress
regulators of gene expression at the post- conditions remain insufficient to support the extreme
transcriptional level. Thousands of such miRNAs are demand of mature RBCs in a short span of time. Bone
identified in various organisms including unicellular marrow produces more number of CFU-E cells from
microorganism and complex multicellular organisms BFU-E cells. This process of generating more number
showing that regulatory miRNA pathway is conserved of BFU-/CFU-E is independent of EPO-mediated
in evolution (Listowski et al., 2013). miRNAs are activities. It is shown that single BFU-E cell can give
defined as a class of small regulatory RNAs that rise to hundreds of CFU-Es in vitro. BFU-E
down-regulates their target genes expression either progenitors may undergo limited self-renewal during
through degradation of mRNA or inhibiting their stress erythropoiesis supporting the survival of
translation (Bartel et al., 2009; Guo et al., 2010). lethally irradiated mice from anemia. Even more,
Erythropoiesis is also regulated by several miRNAs at these BFU-Es can be re-transplanted and protects
various stages which include lineage determination, secondary and tertiary recipients (Harandi et al.,
progenitor proliferation, terminal differentiation of 2010).
erythroid, and enucleation of reticulocytes (Table 2, Under stress conditions, a significant number of
Figure-6). Out of them miR-24, 144, 150, 191, 221, cell division, self-renewal, and differentiation
222, 223, & 451 is reported to have significant roles activities take place which are regulated by various
in human erythropoiesis. miR-150 come into the factors such as microenvironment growth factor/
action at the initial stage and down-regulates Glucocorticoids and Hedgehog signaling along with
expression of Myb gene to inhibit erythroid lineage tissue hypoxia. This is evident from the studies
differentiation (Lu et al., 2008). While miR-24 showing the movement of erythropoietic activities
negatively regulates activin signaling, it down- from the bone marrow to the spleen during stress
regulates the mRNA encoding the Activin receptor- conditions until optimal conditions are restored.
like kinase 4 (ALK4) (Wang et al., 2008). LMO2 is a (McCulloch et al., 1964; Hara et al, 1976). Fetal liver
critical transcription factor which gets up-regulated and spleen-derived stromal cell lines are demonstrated
during erythroid differentiation of CD34- progenitor to inherit better-supporting properties for in vitro fetal
cells and is required for erythroid differentiation, but BFU-E proliferation in comparison to bone marrow
gets down-regulated due to overexpression of miR- stroma cells (Slaper-CortenbachI et al., 1987; Yanai
223 (Felli et al., 2009). During the terminal erythroid et al, 1989; Ohneda et al., 1990). Furthermore, the
progenitor proliferation miR-221 and miR-222 come microenvironment have a significant role in
into action where these miRNAs enhances terminal sustaining erythroid progenitors self-renewal. This is
differentiation by down-regulating the KIT gene that also evident from the mice studies showing resistance
regulates proliferation of erythroid progenitors (Felli to Friend erythroid leukemia virus-induced erythroid
et al., 2005) leukemia (Kreja et al., 1985). A number of attempts
have been made by various researcher groups to
define various regulating factors in spleen and fetal
Category-II: Regulation of Erythropoiesis liver.
under Stress Conditions Another important regulatory factor is the
glucocorticoids (GC) which are released from the
As discussed above the erythropoiesis level is adrenal glands during conditions of stress
several folds enhanced under stress conditions such as erythropoiesis (sepsis/severe trauma). Prednisolone,
hypoxia, chronic anemia, and acute blood loss. This which is a GC analog, is often used to treat the
162 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

patients with the RBC progenitor disorder Diamond- al., 2010). That is how it may have positive effects on
Blackfan anemia. (Flygare et al, 2007; Vlachos et al., a rapid and long-lasting increase in RBC production.
2008). Studies in mice lacking GC receptors or The presence of Hypoxia-induced factor-1 (HIF-
expressing a mutated version GC (deficient in DNA 1) binding sites in the promoter regions of the most of
binding or transactivation) has shown a normal the genes which are induced through GCs mediated
steady-state erythropoiesis but severely impaired signals indicate a link between Hypoxia and GCs
stress erythropoiesis. (Reichardt et al., 1998; Bauer et signaling. It suggests that activation of HIF-1 may
al., 1999). Similarly, GC receptors deficiency resulted enhance or replace the effect of GCs on BFU-E self-
to loss of responsiveness to phenylhydrazine-induced renewal. This is evident from studies showing
hemolysis and failed to increase CFU-E numbers in enhanced production of CFU-E and then erythroblasts
the spleen. approximately through HIF-1 activation by a prolyl
There are reports showing antagonistic effects of hydroxylase inhibitor (Flygare et al., 2011).
p53 activation on GC receptor-induced erythropoiesis.
p53-/-mice shown the rapid growth of CFU-E and
c-Kit/CD34cells that is faster than normal mice in Other Important Issues Related to
response to hypoxic stress in the spleen.(Ganguli et al,
2002).
Ex Vivo RBCs production
It is reported that GCs activity is facilitated
Despite significant progress achieved by
through the induction of expression of Myb, Kit, and
researchers in the field of ex vivo expansion
Lmo2 and inhibition of Gata1 expression (Ganguli et
techniques for blood components during past several
al., 2002; von Lindern et al., 1999). The up-regulated
years, a number of substantial issues remain to be
expression of Kit mRNA is detected as early as 4
solved to ensure the production of manmade blood
hours after GC stimulation of BFU-E cells (Flygare et
components for clinical uses. Altogether these issues
al., 2011). The stress erythropoietic regulation by GCs
may be categorized as (1) initial source material
can be modeled in vitro by growing early erythroid
related issues defining their suitability and proper
progenitors in the presence of SCF, GCs, and Epo
availability for large scale production, (2) quantitative
(Bauer et al., 1999; Flygare et al., 2011; Kolbus et al.,
issues to ensure sufficient production of transfusable
2003; Wessely et al., 1997). These studies reported
units (3) optimal and standardized use of various
that disruption of GC receptor dimerization severely
growth factors in different protocols, (4) biophysical
reduces in vitro proliferation of fetal liver
and biochemical parameters assessment of the
erythroblasts (Reichardt et al., 1998; Bauer et al.,
cultured blood cells to ensure native blood cell-like
1999). This suggests a significant role of GC receptor
properties, (5) vastly defined antigenic profiles to
dimerization in GCs induced RBC production. GCs
avoid any mismatch and associated adverse effects,
dimerization is required for efficient transactivation of
(6) ethnicity, (7) enucleating efficiency of RBCs, (8)
promoters and/or gene repression (Reichardt et al.,
and most importantly production cost
1998; Bauer et al., 1999).
The source materials preferably should be a
GCs induce limited self-renewal of BFU-E cells,
discarded material to reduce the overall production
and not of CFU-E cells or erythroblasts as reported by
cost. To establish a consistent blood manufacturing-
studies using mouse fetal liver BFU-E/CFU-E cells
supplying chain, a regular availability of the source
grown in serum-free conditions along with SCF and
material would be necessary. Apart from that source
IGF-1. These studies have shown protection of BFU-
material should have huge potential to produce
E cells from exhaustion and enhanced formation of
functionally matured cells without eliciting any
CFU-E per BFU-E (Flygare et al., 2011). It is also
immunogenic reactions in the recipients on the
proposed by Lodish and Flygare that increased levels
infusion of the product. One of the most discussed
of GCs likely help in maintaining the earliest
and promising easily available clinical by-product is
erythroid progenitors, increase generation of CFU-E,
umbilical cord blood that is readily available in all the
and may stimulate terminal differentiation (Lodish et
maternity hospitals. The suitability of UCB is also
evident from the facts that significant number of
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 163

transfusion are possible for the rare hemo- reported by using hollow fiber bioreactor technique
globinopathies patients (1%) by using ex vivo (Housler et al., 2012) (Table 3). These reports provide
cultivated RBCs units that can be generated from enough temptations, and it is believed that 3D
UCB (>90ml discarded regularly). It is demonstrated bioreactors with some scaling up designs / method-
that each discarded unit of UCB may give rise to 10 ologies may be a suitable tool to produce 12 units of
75 RBCs products which might be useful in RBCs in 34 weeks of time.
transfusions for rare hemoglobinopathies (Peyrard et The clinical utility of cultured RBCs or other
al., 2011). Another important source may be derived blood components would definitely require the
from Leukopheresis, which is routinely done in development of stringent quality checks to ensure
clinical centers. The cells obtained from leuko- their safety and native-like functional quality. The
reduction buffy coat are demonstrated to have determination of antigenic profiles and hemoglobin
significant amounts of RBCs producing cells contents and identification of physical progenies
(equivalent to umbilical CB) (Zhou et al., 2009). The derived from these culture processes would be of
clinical use was demonstrated by Migliaccio et al. utmost importance. Reports from many laboratories
who shown successful transfusion of CD34+ cells have shown slightly macrocytic nature of the cultured
obtained from a G-CSF mobilized donor (Migliaccio RBCs but normal in size. There are significant level
et al., 2010). Besides, ESCs, and hiPSCs based of -hemoglobin stabilizing proteins (AHSP)
methodology might serve as a potential source for expression, and heterogeneity in the expression of
regular manufacturing processes with the BCL11A and globin genes (Keel et al., 2008). The ex
advancement in technologies. vivo generated erythroblasts express a slightly higher
Since RBCs are the most frequently transfused level of -globin in comparison to native RBCs (Lu et
blood component and are very high in population al., 2008).
(21012 cells/unit) and rich cell density (3106 The number of surface antigens plays vital role in
cells/ml.). As WHO data states >100 million units of maintaining the function of RBCs. Many surface
blood are collected every year, and it remains largely antigens are known for the native mature RBCs. The
insufficient to support the global annual blood unit similar antigenic profile is demonstrated on ex vivo
demands. Obviously, ex vivo expansion of RBCs generated RBCs such as expression of the normal
alone would require a vast number of skilled level of antigens present on both the Ankyrin A
manpower along with huge amounts of essential (GPA, M/N, Ena FS, RhAO, and band 3/4.1 R) and
ingredients. Glycophorin B complex. Similarly, other important
As per a hypothetical estimation, a cell density of surface antigens are also expressed including the urea
5107 cells/ml is critical to producing regularly to transporter, the complement receptor, and receptors
meet this demand. It is also presumed that generation that protect RBCs from complement-mediated lysis.
of a single unit of blood (200 ml) through static Satchwell et al. also reported an appropriate level of
culture methods may need unexpectedly huge amount surface antigens such as band 3 and Rh antigen by ex
of culturing media, growth factors and culturing vivo generated RBCs (Satchwell et al., 2011).
vessels (660 liters medium/ 9500 lab scale 175 cm2 One of the important parameter to determine the
culturing flask) (Timmins and Nielsen, 2009; Zeuner quality/functionality of cultured RBCs is to assess
et al., 2012). Development of automated methods their degree of maturation. Enucleation efficiency of
such as various types of bioreactors has been reported the cultured RBCs is an inevitable requirement for all
in recent past few years. The advancement of their of these production methods. ESCs derived
design, culturing protocols could be helpful for the erythroblasts may yield >60% enucleated RBCs (Lu
researchers to achieve required cell density. Timmins et al., 2008). Whereas, the nucleation efficiency of
et al. have demonstrated the generation of high cell maturing erythroblast from UCB is merely 410% of
density of RBCs in a wave bioreactor (Timmins et al., RBCs produced (Lapillonne et al., 2010). The factors
2011). Similarly, the cell density of 107 cells/ml is regulating enucleation efficiency of RBCs are being
reported in a stirred small scale bioreactor (Ratcliffe explored, and one such important protein is Histone
et al., 2012). A massive yield of 2108 cells/ml was deacetylase (specifically HDAC-2 isoform) which
164 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

regulates chromatin condensation and enucleation as an application of molecular agents may be useful to
reported in mice fetal erythroblast (Chang et al., enhance the enucleation yields. Similarly, enucleation
2011). Interestingly, GCs are shown to function as efficiency may also be improved by using methods of
important negative regulators for these proteins as co -culturing of maturating erythroblast along with
mentioned above, and activation of HDACs through stromal cells.

Table 3. Bioreactors for ex vivo RBC production

S. No. Year Design and Key factors Limitations References


First stirred bioreactor with pH and DO control for Collins et al.,
1 1998 No data on cell maturation
expansion of HSCs Total cells about 400 times 1998
2 2006 Rotating wall vessel bioreactor Complex design Liu et al., 2006
Timmins et al.,
3 2011 High yield up to 500units/ UCB Inconsistent enucleation
2011
15ml minimal scale stirred and monitored Ratcliffe et al.,
5 2012 No data on cell maturation
bioreactor with 6 populations doublings in 10 days 2012
A complex system with poor cell
Custom made hollow fiber bioreactor with
expansion and maturation, with Housler et al.,
6 2012 compartmental Hollow Fiber Capillary Membrane.
only 40% CD235+ cells, Actual 2012
Maximal cell concentration.
size 8ml and 2ml.
Bio-mimetic co-culture platform using hollow fiber Xue et al.,
7 2014 Limited differentiation
bioreactor with 14288 fold expansion 2014

However, co-culture methods are not appreciated immediately achievable goals from the outcomes of
much due to their inherent property to impose the existing research endings may play a major role in
improperly defined potentially immunogenic and/or generating money for the same.
infectious agent and thus can compromise the quality
production of GMP for RBCs. The use of synthetic
drugs e.g., Mifepristone and plasmanate may provide Prospective
a better safe alternate (Miharada et al., 2006).
Besides, other molecules promoting the vesicle Blood components can be produced in limited
trafficking such as vaccuoline may also be used to quantities through different methods and types of
enhance the enucleation efficiency if employed during stem cells available to us. There are many important
phase-3 cultures (Keerthivasan et al., 2010). issues including the quantity and quality of these
The most substantial factor which would define products, and a regular commercial production would
the fate of commercial production of RBCs is cost of only be possible after advancements in the present
production. It has been proposed that by using present culturing technologies in near future.
methodologies available for ex vivo expansion the The studies done so far regarding molecular
approximated cost for RBCs production would lie mechanism of various growth factors and their impact
$800015000 per unit (Timmins and Nielsen, 2009; on the various developmental stages of the
Zeuner et al., 2012). This may look too high if one erythropoiesis have provided profound insights into
compares the hospital cost for leuco-reduced RBC, these processes, and we can regulate both the
which is only $225. Even more, the cost for the rare quantitative and qualitative issues related to RBCs
phenotypically matched unit of RBCs is $7001200 production. Furthermore, the comprehensive
that is significantly low in comparison to the proposed information about the various molecules and
synthetic version of RBCs (Table 4). Further, development of their synthetic alternatives may be
commercial scale production would fetch a huge helpful in finding novel treatments for anemia and
monetary investment. It may not be easy to motivate other RBC disorders. Recent findings in the
financial inflows in the direction of R&D in most of regulation mechanism of -hemoglobin switching
the low-income grade countries. Identification of
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 165

may lead to develop more efficient remedies for different stages of RBC development would enable
related disorders, such as -thalassemia and sickle regular large-scale production of functional RBCs
cell disease (Sankaran et al., 2010). Similarly, from various types of stem cells for clinical blood
advancement in the understanding of different genetic transfusions.
and epigenetic programs which essentially regulate

Table 4. Brief details of approximate cost 1L culture ingredients used in bioreactor

S. No. Parameter Concentration Quantity Approximate Cost in USD


1 Basal Medium (IMDM) - 1L $80
2 Media Supplements
a) Transferrin 330 ug/ml 330 mg $235
b) Insulin 10 ug/ml 10 mg $35
c) Human Serum 5% 50 ml $100
3 Growth Factors
a) Erythropoietin 2I U/ml 2000 IU $2650
b) Interleukin-3 5 ng/ml 5 ug $100
c) Stem Cell Factor 100 ng/ml 100 ug $1500
4 Chemicals
a) Dex/ Hydrocortisone 1 uM 36.2 mg $50

Baek, E. J., Kim, H. S., Kim, J. H., Kim, N. J., and Kim, H. O.
Acknowledgments (2009). Stroma-free mass production of clinical-grade red
blood cells (RBCs) by using poloxamer 188 as an RBC
We are thankful to the Honorable Chairman and survival enhancer. Transfusion 49, 22852295. doi:
the Honorable Vice- Chancellor of the Delhi 10.1111/j.1537-2995.2009.02303.x.
Technological University, Shahbad Daulatpur, Bagnis, C., Chiaroni, J., and Bailly, P. (2011). Elimination of
Bawana road, Delhi-42, for support. Dr. Vimal K. blood group antigens: Hope and reality. Br. J. Haematol.
Singh particularly thanks the Department of Science 152, 392400. doi: 10.1111/j.13652141.2010.08561.x.
Bartel, D. P. (2009). MicroRNAs: target recognition and
and Technology and Indian National Science regulatory functions. Cell 136(2), 215233. doi: 10.1016/
Academy (INSA), INDIA, for the research grant. j.cell.2009.01.002.
Basu, S., Dunn, A., and Ward, A. (2002). G-CSF: function and
modes of action (Review). Int J Mol Med.10 (1), 310.
Bauer, A., Tronche, F., Wessely, O., et al. (1999). The
References glucocorticoid receptor is required for stress
erythropoiesis. Genes Dev. 13(22), 29963002.
Akashi, K., Traver, D., Miyamoto, T., and Weissman, I.L.
Cantor, A. B., and Orkin, S. H. (2002). Transcriptional
(2000). A clonogenic common myeloid progenitor that
regulation of erythropoiesis: an affair involving multiple
gives rise to all myeloid lineages. Nature. 404(6774),
partners. Oncogene. 21(21), 33683376.
193197.
Castro, C. I., and Briceno, J. C. (2010). Perfluorocarbonbased
Ali, A., Auvinen, M. K., and Rautonen, J. (2010). The aging
oxygen carriers: Review of products and trials. Artif.
population poses a global challenge for blood services.
Organs 34, 622634. doi: 10.1111/j.15251594.2009.
Transfusion 50, 584588. doi: 10.1111/j.1537-2995
00944.x.
.2009.02490.x.
Chang, C. J., Mitra, K., Koya, M., Velho, M., Desprat, R.,
Alter, H. J., and Klein, H. G. (2008). The hazards of blood
Lenz, J., et al. (2011). Production of embryonic and fetal-
transfusion in historical perspective. Blood 112, 2617
like red blood cells from human induced pluripotent stem
2626. doi: 10.1182/blood-2008-07-077370.
cells. PLoS ONE 6, 10. doi: 10.1371/journal.pone
Arcasoy, M. O., Amin, K., Chou, S. C., Haroon, Z. A., Varia,
.0025761.
M., and Raleigh, J. A. (2005). Erythropoietin and
Cheng, Y., Wu, W., Kumar, S. A., Yu, D., Deng, W., Tripic,
erythropoietin receptor expression in head and neck
T., et al. (2009). Erythroid GATA1 function revealed by
cancer: relationship to tumor hypoxia. Clinical Cancer
genome-wide analysis of transcription factor occupancy,
Research 11, 2027.
166 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

histone modifications, and mRNA expression. Genome co-culturing with macrophages. Int. J. Hematol. 87, 339
Res. 19(12), 21722184. doi: 10.1101/gr.098921.109. 350. doi: 10.1007/s12185-008-0062-y.
Daniels, G., Castilho, L., Flegel, W. A., Fletcher, A., Garratty, Fujiwara, T., OGeen, H., Keles, S., Blahnik, K., Linnemann,
G., Levene, C., et al. (2009). International society of A. K., Kang, Y.A., et al. (2009). Discovering hemato-
blood transfusion committee on terminology for red blood poietic mechanisms through genomewide analysis of
cell surface antigens: macao report. Vox. Sang. 96, 153 GATA factor chromatin occupancy. Mol Cell. 36(4), 667
156. doi: 10.1111/j.1423-0410.2008.01133.x. 681. doi: 10.1016/j.molcel.2009.11.001.
Department of Health and Human Services. (2010). The 2009 Ganguli, G., Back, J., Sengupta, S., Wasylyk, B. (2002). The
National Blood Collection and Utilization Survey Report. p53 tumour suppressor inhibits glucocorticoidinduced
Washington, DC: DHHS. Available online at: http://www. proliferation of erythroid progenitors. EMBO Rep. 3(6),
aabb.org/programs/biovigilance/nbcus/Documents/09- 569574.
nbcus-report.pdf (Accessed May 4, 2014). Giarratana, M. C., Rouard, H., Dumont, A., Kiger, L.,
Department of Health and Human Services. (2013). The 2011 Safeukui, I., Le Pennec, P. Y., et al. (2011). Proof of
National Blood Collection and Utilization Survey Report. principle for transfusion of in vitro-generated red blood
Washington, DC: DHHS. Available online at: http://www. cells. Blood 118, 50715079. doi: 10.1182/blood-2011-
aabb.org/programs/biovigilance/nbcus/Documents/11- 06-362038.
nbcus-report.pdf (Accessed May 4, 2014). Gregory, C. J., and Eaves, A. C. (1978). Three stages of
Drouet, M., Mathieu, J., Grenier, N., Multon, E., Sotto, J. J., erythropoietic progenitor cell differentiation distinguished
and Herodin, F. (1999). The reduction of in vitro by a number of physical and biologic properties. Blood
radiation-induced Fas-related apoptosis in CD34+ 51, 527537.
progenitor cells by SCF, FLT-3 ligand, TPO, and IL-3 in Guo, H., Ingolia, N. T., Weissman, J. S., and Bartel, D. P.
combination resulted in CD34+ cell proliferation and (2010). Mammalian microRNAs predominantly act to
differentiation. Stem Cells.17 (5), 273285. decrease target mRNA levels. Nature. 466(7308), 835
Elliott, S., Pham, E., and Macdougall, I.C. (2008). 840. doi: 10.1038/nature09267.
Erythropoietin: a common mechanism of action. Exp Hall, M. A., Curtis, D. J., Metcalf, D., Elefanty, A. G., Sourris,
Hematol. 36(12), 15731584. doi: 10.1016/j.exphem.2008 K., Robb, L., et al. (2003). The critical regulator of
.08.003. embryonic hematopoiesis, SCL, is vital in the adult for
Ende, M., and Ende, N. (1972). Hematopoietic transplantation megakaryopoiesis, erythropoiesis, and lineage choice in
by means of fetal (cord) blood. A new method. Va. Med. CFUS12. Proc Natl Acad Sci USA 100, 992997.
Mon. 99, 276280. Hara, H., Ogawa, M. (1976). Erthropoietic precursors in mice
Fawcett, D.W., Bloom & Fawcett (1997) Concise Histology with phenylhydrazine-induced anemia. Am J Hematol.
(1st ed). Oxford, United Kingdom:Arnold Publishers. 1(4), 453458.
Felli, N., Fontana, L., Pelosi, E., Botta, R., Bonci, D., Harandi, O.F., Hedge, S., Wu, D.C., McKeone, D., Paulson,
Facchiano, F., et al. (2005) MicroRNAs 221 and 222 R.F. (2010). Murine erythroid short-term radioprotection
inhibit normal erythropoiesis and erythroleukemic cell requires a BMP4-dependent, self-renewing population of
growth via kit receptor downmodulation. Proc Natl Acad stress erythroid progenitors. J Clin Invest. 120(12), 4507
Sci U S A. 102(50), 18081-18086. 4519.
Felli, N., Pedini, F., Romania, P., Biffoni, M., Morsilli, O., Hegde, S., Lenox, L.E., Lariviere, A., et al. (2007). An intronic
Castelli, G., et al. (2009). MicroRNA 223- dependent sequence mutated in flexed-tail mice regulates splicing of
expression of LMO2 regulates normal erythropoiesis. Smad5. Mamm Genome. 18(12), 852860.
Haematologica. 94(4), 479486. doi: 10.3324/haema Henkel-Honke, T., and Oleck, M. (2007). Artificial oxygen
tol.2008.002345. carriers: a current review. AANA. J. 75, 205211.
Flygare, J., Karlsson, S. (2007). Diamond-Blackfan anemia: Hiroyama, T., Miharada, K., Sudo, K., Danjo, I., Aoki, N., and
erythropoiesis lost in translation. Blood. 109(8), 3152 Nakamura, Y. (2008). Establishment of mouse embryonic
3154. stem cell-derived erythroid progenitor cell lines able to
Flygare, J., Rayon, Estrada, V., Shin, C., Gupta, S., Lodish, produce functional red blood cells. PLoS ONE 6:3. doi:
H.F., (2011). HIF-1alpha synergizes with glucocorticoids 10.1371/journal.pone.0001544.
to promote BFU-E progenitor self-renewal. Blood. Housler, G. J., Miki, T., Schmelzer, E., Pekor, C., Zhang, X.,
117(12), 34353444. Kang, L., et al. (2012). Compartmental hollow fiber
Fuchs, O., Simakova, O., Klener, P., et al. (2002). Inhibition of capillary membrane-based bioreactor technology for in
Smad5 in human hematopoietic progenitors blocks vitro studies on red blood cell lineage direction of
erythroid differentiation induced by BMP4. Blood Cells hematopoietic stem cells. Tissue Eng. Part. C Methods
Mol Dis. 28(2), 221233. 18, 133142. doi: 10.1089/ten.tec. 2011.0305.
Fujimi, A., Matsunaga, T., Kobune, M., Kawano, Y., Nagaya, Inaba, K., Inaba, M., Romani, N., Aya, H., Deguchi, M.,
T., Tanaka, I., et al. (2008).Exvivolarge-scalegeneration Ikehara, S., et al. (1992). RM. Generation of large
of human red blood cells from cord blood CD34+ cells by numbers of dendritic cells from mouse bone marrow
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 167

cultures supplemented with granulocyte/macrophage primary human erythroid progenitors. Blood 105, 8594.
colony-stimulating factor. J Exp Med. 176(6), 1693702. doi: 10.1182/blood-2004-03-1002.
James, W. F. (2003). Erythropoietin: Physiology and Lecuyer, E., Herblot, S., Saint-Denis, M., Martin, R., Begley,
Pharmacology Update, Exp Biol Med (Maywood) 228, 1. C. G., Porcher, C., et al. (2002). The SCL complex
Jiang, X. S., Chai, C., Zhang, Y., Zhuo, R. X., Mao, H. Q., and regulates c-kit expression in hematopoietic cells through
Leong, K. W. (2006). Surface immobilization of adhesion functional interaction with Sp1. Blood 100, 24302440.
peptides on the substrate for ex vivo expansion of Lenox, L.E., Perry, J.M., Paulson, R.F. (2005). BMP4 and
cryopreserved umbilical cord blood CD34+ cells. Madh5 regulate the erythroid response to acute anemia.
Biomaterials. 27(13), 272332. Blood. 105(7), 27412748.
Kassouf, M. T., Hughes, J. R., Taylor, S., McGowan, S. J., Lindern, M., Schmidt, U., and Beug, H. (2004). Control of
Soneji, S., Green, A. L., et al. (2010). Genomewide erythropoiesis by erythropoietin and stem cell factor: a
identification of TAL1s functional targets: insights into novel role for Bruton's tyrosine kinase. Cell Cycle. 3(7),
its mechanisms of action in primary erythroid cells. 876879.
Genome Res. 20(8), 10641083, doi: 10.1101/gr. Listowski, M. A., Heger, E., Bogusawska, D. M., Machnicka,
104935.110. B., Kuliczkowski, K., Leluk, J., et al. (2013). microRNAs:
Kawada, H., Ando, K., Tsuji, T., Shimakura, Y., Nakamura, fine tuning of erythropoiesis. Cell Mol Biol Lett. 18(1),
Y., Chargui, J., et al. (1999). Rapid ex vivo expansion of 3446. doi: 10.2478/s11658-012-0038-z.
human umbilical cord hematopoietic progenitors using a Lodish, H., Flygare, J., and Chou, S. (2010). From stem cell to
novel culture system. Exp Hematol. 27(5), 90415. erythroblast: regulation of red cell production at multiple
Keel,S.B.,Doty,R.T.,Yang,Z.,Quigley,J.G.,Chen,J.,Knoblaugh, levels by multiple hormones. IUBMBLife 62, 492496.
S.,etal.(2008). A heme export protein is required for red doi: 10.1002/iub.322.
blood cell differentiation and iron homeosiasis. Science Love, P. E., Warzecha, C., and Li, L. (2014). Ldb1 complexes:
319, 825828. doi: 10.1126/science.1151133. the new master regulators of erythroid gene transcription.
Keerthivasan, G., Small, S., Liu, H., Wickrema, A., and Trends Genet. 30(1), 19. doi: 10.1016/j.tig.2013.10.001.
Crispino, J.D. (2010).Vesicle trafficking plays a novel Lu, J., Guo, S., and Ebert, B. L., et al. (2008). MicroRNA-
role in erythroblast enucleation. Blood 116, 33313340. mediated control of cell fate in megakaryocyte-
doi: 10.1182/blood-2010-03-277426. erythrocyte progenitors. Dev Cell. 14(6), 843853. doi:
Kolbus, A., Blazquez-Domingo, M., Carotta, S., et al. (2003). 10.1016/j.devcel.2008.03.012.
Cooperative signaling between cytokine receptors and the Lu, S. J., Feng, Q., Park, J. S., Vida, L., Lee, B. S.,
glucocorticoid receptor in the expansion of erythroid Strausbauch, M., et al. (2008). Biologic properties and
progenitors: molecular analysis by expression profiling. enucleation of red blood cells from human embryonic
Blood. 102(9), 31363146. stem cells. Blood 112, 44754484. doi: 10.1182/blood-
Korpelainen, E. I., Gamble, J. R., Vadas, M. A., and Lopez, A. 2008-05-157198.
F. (1996). IL-3 receptor expression, regulation and Lugus, J.J., Chung,Y.S., Mills, J.C. et al. (2007). GATA2
function in cells of the vasculature. Immunol Cell Biol. functions at multiple steps in hemangioblast development
74(1), 17. and differentiation. Development. 134(2), 393405.
Koury, M.J. and Bondurant, M.C. (1990). Erythropoietin McCulloch, E.A., Siminovitch, L., Till, J.E. (1964).
retards DNA breakdown and prevents programmed death Spleencolony formation in anemic mice of genotype Ww.
in erythroid progenitor cells. Science. 248(4953), 378 Science. 144, 844846.
381. Metcalf, D., and Nicola, N. A. (1995). The hematopoietic
Kreja, L., Seidel, H.J. (1985). On the role of the spleen in colony stimulating factors. New York, NY: Cambridge
Friend virus (F-MuLV-P) erythroleukemia. Exp Hematol. University Press.
13(7), 623628. Michael, R. T., and Perkins, C. A. (2010). KLF1 Directly
Kuo, Y. Y., and Chang, Z. F. (2007). GATA-1 and Gfi-1B Coordinates Almost All Aspects of Terminal Erythroid
interplay to regulate Bcl-xL transcription. Mol Cell Biol. Differentiation. IUBMB Life, 62(12), 886890. doi:
27, 42614272. 10.1002/iub.404.
Lapillonne, H., Kobari, L., Mazurier, C., Tropel, P., Migliaccio, A. R., Masselli, E., Varricchio, L., and Whitsett, C.
Giarratana, M. C., Cleon, I. Z., et al. (2010). Red blood (2012). Ex-vivo expansion of red blood cells: how real for
cell generation from human induced pluripotent stem transfusion in humans? Blood Rev. 26, 8195. doi:
cells: perspectives for transfusion medicine. 10.1016/j.blre.2011.11.002.
Haematologica 95, 16511659. doi: 10.3324/haematol. Migliaccio, G., Sanchez, M., Masiello, F., Tirelli, V.,
2010.023556. Varricchio, L., Whitsett, C., et al. (2010). Humanized
Leberbauer, C., Boulme, F., Unfried, G., Huber, J., Beug, H., culture medium for clinical expansion of human
and Mullner, E. W. (2005). Different steroids coregulate erythroblasts. Cell Transplant. 19, 453469. doi: 10.3727/
long-term expansion versus terminal differentiation in 096368909X485049.
168 Vimal Kishor Singh, Abhishek Saini, Manisha Kalsan et al.

Miharada, K., Hiroyama, T., Sudo, K., Nagasawa, T., and culture. J. Biotechnol. 161, 387390. doi: 10.1016/
Nakamura, Y. (2006). Efficient enucleation of j.jbiotec.2012. 06.025.
erythroblasts differentiated in vitro from hematopoietic Ravet, E., Reynaud, D., Titeux, M., Izac, B., Fichelson, S.,
stem and progenitor cells. Nat. Biotechnol. 24, 1255 Romo, P. H., et al. (2004). Characterization of DNA-
1256. doi: 10.1038/nbt1245. binding-dependent and - independent functions of
Millot, S., Andrieu, V., Letteron, P. et al. (2010). SCL/TAL1 during human erythropoiesis. Blood 103,
Erythropoietin stimulates spleen BMP4-dependent stress 33263335.
erythropoiesis and partially corrects anemia in a mouse Reichardt, H.M., Kaestner, K.H., Tuckermann, J. et al. (1998).
model of generalized inflammation. Blood. 116(26), DNA binding of the glucocorticoid receptor is not
60726081. essential for survival. Cell. 93(4), 531541.
Molineux, G., Foote, M., Elliott, S. (2009). Erythropoiesis and Rosas, M., Gordon, S., and Taylor, P. R. (2007).
Erythropoietins (2nd ed). Basel, Switzerland: Birkhauser. Characterization of the expression and function of the
Munugalavadla, V., and Kapur, R. (2005). The role of c-Kit GM-CSF receptor -chain in mice. Eur J Immunol. 37(9),
and erythropoietin receptor in erythropoiesis. Crit Rev 25182528.
Oncol Hematol. 54(1), 6375. Sankaran, V.G., Nathan, D.G. (2010). Reversing the
Natanson, C., Kern, S. J., Lurie, P., Banks, S. M., and Wolfe, hemoglobin switch. N Engl J Med. 363(23), 22582260.
S. M. (2008). Cell free hemoglobin-based blood Satchwell, T. J., Bell, A. J., Pellegrin, S., Kuziq, S., Ridgwell,
substitutes and risk of myocardial infarction and death: a K., Daniels, G., et al. (2011). Critical band 3 multiprotein
meta-analysis. J. Am. Med. Assoc. 299, 23042312. doi: complex interactions establish early during human
10.1001/jama.299.19.jrv80007. erythropoiesis. Blood 118, 182191. doi: 10.1182/blood-
Neildez-Nguyen, T.M., Wajcman, H., Marden, M. C., 201010-314187.
Bensidhoum, M., Moncollin, V., Giarratana, M. C., et al. Silverman, T. A., and Weiskopf, R. B. (2009).
(2002). Human erythroid cells produced cx vivo at large Hemoglobinbased oxygen carriers: Current status and
scale differentiate into red blood cells in vivo. Nat. future directions. Transfusion 49, 24952515. doi:
Biotechnol. 20, 467472. doi: 10.1038/nbt0502-467. 10.1111/j.1537-2995.2009.02356.x.
Ohneda, O., Yanai, N., Obinata, M. (1990). Microenvironment Singbrant, S., Karlsson, G., Ehinger, M. et al. (2010).
created by stromal cells is essential for a rapid expansion Canonical BMP signaling is dispensable for
of erythroid cells in mouse fetal liver. Development. hematopoietic stem cell function in both adult and fetal
110(2), 379384. liver hematopoiesis, but essential to preserve colon
Orkin, S. H. (1992). GATA-binding transcription factors in architecture. Blood. 115(23), 46894698.
hematopoietic cells. Blood 80, 575581. Singbrant, S., Moody, J.L., Blank, U. et al. (2006). Smad5 is
Orkin, S. H. (2000). Diversification of hematopoietic stem dispensable for adult murine hematopoiesis. Blood.
cells to specific lineages. Nat Rev Genet 1, 5764. 108(12), 37073712.
Papetti, M., Wontakal, S.N., Stopka, T., and Skoultchi, A. I. Singh, V.K., Saini, A., Tsuji, K., Sharma, P.B., and Chandra,
(2010). GATA-1 directly regulates p21 gene expression R. (2014). Manufacturing blood ex vivo: a futuristic
during erythroid differentiation. Cell Cycle. 9(10), 1972 approach to deal with the supply and safety concerns.
80. Front. Cell Dev. Biol. doi: 10.3389/fcell.2014.00026.
Perry, J.M., Harandi, O.F., Porayette, P., Hegde, S., Kannan, Slaper-Cortenbach, I., Ploemacher, R., Lowenberg, B. (1987).
A.K., Paulson, R.F. (2009). Maintenance of the BMP4- Different stimulative effects of human bone marrow and
dependent stress erythropoiesis pathway in the murine fetal liver stromal cells on erythropoiesis in long-term
spleen requires hedgehog signaling. Blood. 113(4), 911 culture. Blood. 69(1), 135139.
918. Stanley, E. R., Berg, K. L, Einstein, D. B., Lee, P. S., Pixley,
Peyrard, T., Bardiaux, L., Krause, C., Kobari, L., Lapillonne, F. J., Wang, Y., et al. (1997). Biology and action of the
H., Andreu, G., et al. (2011). Banking of pluripotent adult colony--stimulating factor-1. Mol. Reprod. Dev. 46 (1), 4
stem cells as an unlimited source for red blood cell 10.
production; Potential applications for alloimmunized Subramanian, A., Hegde, S., Porayette, P., Yon, M., Hankey,
patients and rare blood challenges. Transfus. Med. Rev. P., Paulson, R.F. (2008). Friend virus utilizes the BMP4-
25, 206216. doi: 10.1016/j.tmrv.2011.01.002. dependent stress erythropoiesis pathway to induce
Pharr, P. N., Hofbauer, A., Worthington, R. E., and Longmore, erythroleukemia. J Virol. 82(1), 382393.
G. D. (2000). Residual erythroid progenitors in W/W Szabo, E., Rampalli, S., Risueno, R. M., Schnerch, A.,
mice respond to erythropoietin in the absence of steel Mitchell, R., Fiebig-Comyn, A., et al. (2010). Direct
factor signals. Int J Hematol. 72(2), 17885. conversion of human fibroblasts to multilineage blood
Ratcliffe, E1., Glen, K. E., Workman, V. L., Stacey, A. J., and progenitors. Nature 468, 521526. doi: 10.1038
Thomas, R. J. (2012). A novel automated bioreactor for /nature09591
scalable process optimisation of haematopoietic stem cell Timmins, N. E., and Nielsen, L. K. (2009). Blood cell
manufacture: current methods and future challenges.
Stage-Specific Regulation of Erythropoiesis and Its Implications in Ex-Vivo RBCs Generation 169

Trends Biotechnol. 27, 415422. doi: 10.1016/j.tibtech Wessely, O., Deiner, E.M., Beug, H., von Lindern, M. (1997).
.2009.03.008. The glucocorticoid receptor is a key regulator of the
Timmins, N. E., Athanasas, S., Gnther, M., Buntine, P., and decision between self-renewal and differentiation in
Nielsen, L. K. (2011). Ultrahigh-yield manufacture of red erythroid progenitors. EMBO J. 16(2), 267280.
blood cells from hematopoietic stem cells. Tissue Eng. Winslow, R. M. (2006). Current status of oxygen carriers
Part. C Methods 17, 11311137. doi: 10.1089/ten (blood substitutes): 2006. Vox Sang. 91, 102110. doi:
.tec.2011.0207. 10.1111/j.1423-0410.2006.00789.x.
Tripic, T., Deng, W., Cheng, Y., Zhang, Y., Vakoc, C. R., World Health Organization. (2011). Global Database on Blood
Gregory, G. D., et al. (2009). SCL and associated proteins Safety: Summary Report 2011. Available online at:
distinguish active from repressive GATA transcription http://www.who.int/bloodsafety/global_database/GDBS_
factor complexes. Blood 113(10), 21912201. doi: Summary_Report_2011.pdf (Accessed May 4, 2014).
10.1182/blood-2008-07-169417. Wu, D.C., Paulson, R.F. (2005). Hypoxia regulates BMP4
U.S. Census Bureau. (2004). Global Population Composition. expression in the murine spleen during the recovery from
Available online at: http://www.census.gov/population/ acute anemia. PLoS One. 5(6), e11303.
international/files/wp02/wp-02004.pdf (Accessed Yanai, N., Matsuya, Y., Obinata, M. (1989). Spleen stromal
September 14, 2012). cell lines selectively support erythroid colony formation.
Vlachos A., Ball, S., Dahl, N. et al. (2008). Diagnosing and Blood. 74(7), 23912397.
treating Diamond Blackfan anaemia: results of an Yu, M., Riva, L., Xie, H., Schindler, Y., Moran, T.B., Cheng,
international clinical consensus conference. Br J y., et al. (2009). Insights into GATA-1- mediated gene
Haematol. 142(6), 859876. activation versus repression via genome-wide chromatin
VonLindern, M., Zauner, W., Mellitzer, G. et al. (1999). The occupancy analysis. Mol Cell. 36(4), 682695. doi:
glucocorticoid receptor cooperates with the erythropoietin 10.1016/j.molcel.2009.11.002.
receptor and c-Kit to enhance and sustain proliferation of Zeuner, A., Martelli, F., Vaglio, S., Federici, G., Whitsett, C.,
erythroid progenitors in vitro. Blood. 94(2), 550559. and Migliaccio, A. R. (2012). Concise review: stem cell-
Wadman, I. A., Osada, H., Grutz, G. G., Agulnick, A. D., derived erythrocytes as upcoming players in blood trans-
Westphal, H., Forster, A., et al. (1997). The LIM-only fusion. Stem Cells 30, 15871596. doi: 10.1002/
protein Lmo2 is a bridging molecule assembling an stem.1136.
erythroid, DNA-binding complex which includes the Zhou, H., Wu, S., Joo, J. Y., Zhu, S., Han, D. W., Lin, T., et al.
TAL1, E47, GATA-1 and Ldb1/NLI proteins. EMBO J (2009). Generation of induced pluripotent stem cells using
16, 31453157. recombinant proteins. Cell Stem Cell 4, 381384. doi:
Wang, Q., Huang, Z., Xue, H., Jin, C., Ju, X. L., Han, J. D., et 10.1016/j.stem.2009.04.005.
al. (2008). MicroRNA miR-24 inhibits erythropoiesis by
targeting activin type I receptor ALK4. Blood. 111(2),
588595.

Das könnte Ihnen auch gefallen