Blood Reviews 24 (2010) 27–37
Contents lists available at ScienceDirect
Blood Reviews
journal homepage: www.elsevier.com/locate/blre
REVIEW
Autologous blood cell therapies from pluripotent stem cells
Claudia Lengerke a,*, George Q. Daley b,c,d,e,f,
a
Division of Hematology and Oncology, University of Tuebingen Medical Center II, 72076 Tuebingen, Germany
Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital, Boston, MA 02115, USA
c
Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
d
Manton Center for Orphan Disease Research, Boston, MA 02115, USA
e
Harvard Stem Cell Institute, Cambridge, MA 02138, USA
f
Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
b
a r t i c l e
i n f o
Keywords:
Human embryonic stem cells
ESC
Induced pluripotent stem cells
iPS
Differentiation
Hematopoietic stem cells
Blood
Transplantation
Autologous
Isogenic
Erythrocytes
NK cells
Neutrophils
Lymphocytes
s u m m a r y
The discovery of human embryonic stem cells (hESCs) raised promises for a universal resource for cell
based therapies in regenerative medicine. Recently, fast-paced progress has been made towards the generation of pluripotent stem cells (PSCs) amenable for clinical applications, culminating in reprogramming
of adult somatic cells to autologous PSCs that can be indefinitely expanded in vitro. However, besides the
efficient generation of bona fide, clinically safe PSCs (e.g., without the use of oncoproteins and gene transfer based on viruses inserting randomly into the genome), a major challenge in the field remains how to
efficiently differentiate PSCs to specific lineages and how to select cells that will function normally upon
transplantation in adults. In this review, we analyse the in vitro differentiation potential of PSCs to the
hematopoietic lineage by discussing blood cell types that can be currently obtained, limitations in derivation of adult-type HSCs and prospects for clinical application of PSCs-derived blood cells.
Ó 2009 Elsevier Ltd. All rights reserved.
Introduction
Studies on pluripotent stem cells (PSCs) started almost three
decades ago with the discovery of mouse embryonic stem cells
(mESCs) by Martin1 and Evans.2 mESCs can differentiate into every
tissue of the adult body: when reinjected in the developing embryo, mESCs chimaerize all tissues, including the germ line. Over
the last years, ESC technology has been extensively used for genetic manipulation of the mouse genome and creation of mutant
mouse strains, rendering enormously valuable insights into genetic
regulation of tissue function and disease pathogenesis. The discovery of human embryonic stem cells (hESCs) in 19983 opened up
exciting prospects for the use of ESCs in regenerative medicine.
Several organs of the adult are dependent on a stem cell pool for
maintenance, and malignant or degenerative disorders affecting
* Corresponding author. Address: Division of Hematology and Oncology, University of Tuebingen Medical Center II, Otfried-Mueller-Str. 10, 72076 Tuebingen,
Germany. Tel.: +49 7071 2982912; fax: +49 7071 294524.
E-mail addresses: claudia.lengerke@med.uni-tuebingen.de (C. Lengerke), george.daley@childrens.harvard.edu (G.Q. Daley).
Address: Division of Hematology/Oncology, Children’s Hospital Boston, 300
Longwood Avenue, Boston, MA 02115, USA. Tel.: +1 617 919 2015; fax: +1 617 730
0222.
0268-960X/$ - see front matter Ó 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.blre.2009.10.001
this cellular compartment might be treated with stem cell replacement therapies. The most prominent example is the hematopoietic
system, where transplantation of hematopoietic stem cells (HSCs)4
is a well-established clinical tool in the treatment of malignant
(e.g., leukemia and lymphoma) or genetic blood diseases (e.g. Fanconi’s anemia, immunodeficiency and hemoglobinopathy). More
recently, stem cells have been identified in several other tissues
of the adult (e.g., skin, gut, nervous system, lung, and mammary
gland). However, adult stem cells are rare, and despite attempts
to drive ex vivo expansion, adult stem cells including HSCs remain
difficult to maintain and proliferate in culture. In contrast, ESCs can
be indefinitely expanded in an undifferentiated state, providing
limitless amounts of cellular material. Advances in the generation
of patient-specific PSCs5 open up prospects for the autologous cellular therapies that would lack immune rejection. However, the directed differentiation of PSCs into tissues of interest remains
challenging. Despite intensive efforts, currently available in vitro
differentiation protocols offer limited recapitulation of embryonic
development and obtaining adult-type tissue progenitors that will
function normally upon transplantation remains difficult. In this
review, we discuss possible sources of histocompatible PSCs, analyse in vitro blood differentiation from such pluripotent cells, and
discuss prospects for therapeutical applications.
28
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
Genetically customized grafts from pluripotent stem cells
Hematopoietic stem cell transplantation (HSCT) is the best
established clinical cellular replacement therapy, dating back to
1957 when Thomas and colleagues first reported intravenous
infusions of bone marrow in patients receiving radiation and chemotherapy.4 In the ensuing decades transplantation of allogeneic
HLA-matched bone marrow or mobilized peripheral blood CD34+
cells has become the standard therapy for patients suffering from
a variety of malignant or genetic disorders of the hematopoietic
cell compartment. However, allogeneic HSCT is accompanied by
significant morbidity and mortality related to graft rejection,
acute and chronic graft-versus-host disease (GvHD), as well as
infections occurring during the transition period before transplanted HSCs take over the blood cell function. Autologous HSCT,
in which a patient’s own stem cells are harvested prior to highdose chemotherapy, is less toxic because there is no GvHD and
more rapid engraftment translates into lower rates of infectious
complications. However, in patients with genetic conditions such
as sickle-cell anemia and thalassemia, autologous therapies
necessitate correction of the genetic defect by gene therapy in
the patient’s HSCs, which is cumbersome due to the challenges
of maintaining HSCs in culture, the intrinsic difficulties of
expressing genes in HSCs, and the risk of insertional mutagenesis
after gene transfer with viral vectors.6 In contrast, generating patient’s own PSCs, and using for example homologous recombination to correct genetic defects prior to differentiation into
transplantable HSCs promises to overcome caveats of conventional HSCT therapies.
Classically obtained ESCs3 would face immune barriers when
transplanted into (genetically non-identical) hosts. While ESCs
themselves express only low levels of MHC antigens, these levels
increase strongly during differentiation,7 and grafts composed of
ESC-derived progeny would provoke immune reactions and face
rejection upon transplantation in a genetically mismatched host.
Thus, much effort has been invested to generate histocompatible
PSCs. Early work by Briggs and Gurdon in the 1950s8 and
1960s9,10 demonstrated that replacing the nucleus of frog oocytes
with nuclei from somatic cells enables development of organisms
expressing the genetic information of the somatic cell donor. This
principle has been successfully applied in some mammalian species where nuclear transfer (NT)-embryos have been used to derive
ESC lines. NT-ESCs are isogenic with the somatic cell donor, and
thus a source of histocompatible transplant tissue. Rideout and colleagues performed a proof of principle experiment in an immunodeficiency mouse model, showing that such cells can be used for
treatment of genetic disease: NT-ESCs were generated from
Rag2-/- mice; the genetic defect was corrected by homologous
recombination; and the resulting ESCs differentiated in vitro into
repopulating HSCs11 capable of restoring the immune function
upon transplantation into Rag2-/- mice.12
Nuclear transfer is an elegant method for the generation of
isogenic cellular products,13 but the downside of this procedure
is its very low efficiency. To our knowledge, derivation of human
NT-ESCs has not yet succeeded, although there is one report on
successful generation of human blastocysts via NT14 (reviewed
in15). Human NT faces ethical concerns, and is further burdened
by the high numbers of human oocytes that are needed as recipient cells. A more efficient method rendering histocompatible PSCs
is direct oocyte activation in a process called parthenogenesis.16
Parthenogenetic cells are genetically similar, but not identical to
the oocyte donor. Tissue products from parthenogenetic sources
may be homozygous,17,18 and thus susceptible to rejection
through natural killer cells which recognize the missing antigens
upon transplantation into the oocyte donor. However, by analysing high numbers of mouse parthenogenetic ESC lines derived in
our laboratory, we found a surprisingly high degree of histocompatibility due to heterozygosity at the MHC loci, based on early
recombination events during oocyte maturation.16 However, histocompatible parthenogenetic ESCs would be only available for
women capable of oocyte donation, and further analysis should
inform whether imprinting aspects biases their differentiation
into specific tissues.
During NT somatic cell nuclei are reprogrammed by the ooplasm. Significant effort has been expended to identify the mechanisms underlying these processes, in an attempt to replace the
ooplasm effect by defined factors. In 2006, Takahashi and Yamanaka presented ground-breaking results from a screen on a mini-library of embryonic factors for their effect on somatic cells:19 While
single factors were ineffective, combinatorial retroviral transduction with four defined factors known from ESC biology (Oct3/4,
Sox2, c-Myc and Klf4) was able to reprogram mouse fibroblasts to
cells that resembled ESCs both functionally and molecularly.19
The reprogrammed cells could be expanded as cell lines and were
termed as induced pluripotent stem (iPS) cells. Shortly after the
initial report, three independent research groups replicated these
results with human cells.20–22 During the last year and a half, a
multitude of studies from numerous research groups has reported
derivation of reprogrammed PSCs in mouse and human, using different somatic cells as starting populations (e.g. liver and stomach
cells,23 neural stem cells,24,25 pancreatic beta cells,26 B-lymphocytes27 and most recently, human mobilized CD34+ cells from
peripheral blood28), transduction with different embryonic factors,
treatment with histone deacetylase inhibitors29 or small molecules,30 in an attempt to develop protocols devoid of oncoproteins
and integrating viruses31–34 (reviewed in15) that can be translated
into clinical application.
Pluripotent stem cells have also been isolated from mouse and
human embryonic gonads35,36 and more recently from postnatal
testis.37–40 Gonads-derived cells have been shown to form derivatives of all the three germ layers and differentiate into hematopoietic cells.41 To our knowledge, a careful analysis of their potential
to generate distinct blood progenitors and a comparison with pluripotent stem cells from other sources is currently missing. Origin
and characteristics of currently available pluripotent stem cells are
summarized in Table 1.
Challenges of pluripotent stem cells in vitro differentiation
ESCs transplanted into immunodeficient murine recipients form
teratomas, demonstrating their pluripotency. Thus, to obtain specific transplantable tissues, PSCs need to be predifferentiated
in vitro. When removed from the specific culture conditions that
sustain their self-renewal, ESCs spontaneously form cystic structures termed as embryoid bodies (EBs) that contain derivatives of
the three germ layers, including blood cells.42 The presence of serum in the differentiation medium provides a mixture of growth
factors that allows the development of several lineages. This procedure clearly demonstrates the in vitro developmental potency of
ESCs, but has some important disadvantages: (1) the efficiency of
differentiation into specific lineages is highly variable; (2) selection
for the cells of interest (e.g. by surface antigens) is required prior to
transplantation; last but not least (3) the presence of bovine serum
hampers clinical applications requiring protocols free of contaminating animal products.
To drive tissue formation from ESCs and to progress towards directed differentiation protocols using defined serum-free conditions, it is essential to follow a developmental biology approach
(reviewed in43,44). From a developmental standpoint, ESCs are the
equivalent of early epiblast cells and recapitulate in vitro aspects
of early embryogenesis: guided by morphogens (e.g. Wnt, TGF-beta,
Table 1
Origin, properties and hematopoietic potency of different pluripotent stem cells.
Origin
Chimera formation
Hematopoietic potency
HLA-status vs recipients
Key references
Murine ES cells
Blastocyst
Somatic and germline
contribution
Genetically identical (inbred mouse strains)
1,2,42,11
Human ES cells
Blastocyst
Not tested
Genetically unique (HLA-match through banks)
3,50,56,77–
79,104,129
Murine parthenogenetic ES
cells
Oocyte
Somatic contribution
Histocompatible through recombination at MHCloci
16,89
Human parthenogenic ES cells
Murine SCNT ES cells
Oocyte
Somatic cell nucleus,
oocyte
Not tested
Somatic and germline
contribution
Primitive and definitive hematopoiesis
Repopulating HSC (with ectopic
HoxB4)
Primitive and definitive erythropoiesis
Multilineage
hematopoietic
progenitors
Derivation of HSC unclear
Primitive and definitive hematopoiesis
Repopulating HSC (with ectopic
HoxB4)
Not tested
Primitive and definitive hematopoiesis
Repopulating HSC (with ectopic
HoxB4)
Complete HLA-matched or haploidentical
Genetically identical
17,18
12,13
Human SCNT ES cells
Murine iPS cells
Derivation has not been reported
Somatic cells
Somatic and germline
contribution
Genetically identical
14
19,49,51
Human iPS cells
Somatic cells
Not tested
Murine EG cells
Embryonic gonad
Human EG cells
Murine mGS cells
Human mGs cells
Embryonic gonad
Postnatal testis
Postnatal testis
somatic and germline
contribution
Not tested
Somatic contribution
Not tested
Primitive and definitive hematopoiesis
Repopulating HSC (with ectopic
HoxB4)
Primitive and definitive erythropoiesis
Multilineage
hematopoietic
progenitors
Derivation of HSC unclear
Not tested
Genetically identical
5,20–22,54
Genetically identical
35,41
Not tested
Not tested
Not tested
Genetically non-identical
Genetically identical
Genetically identical
36
37–39
40
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
Cell line
Mouse and human pluripotent stem cells of different origins can differentiate into blood cells, derivation of engraftable hematopoietic stem cells has been demonstrated for the majority of mouse pluripotent stem cells, yet remains
cumbersome from human cells.
Abbreviations: ES, embryonic stem; SCNT, somatic cell nuclear transfer; iPS, induced pluripotent stem; EG, embryonic germ; GS, germline stem.
29
30
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
Activin and BMP). ESCs progress through a primitive streak-like
stage before forming the three germ layers, endoderm, ectoderm
and mesoderm,45 which is the layer giving rise to blood. While
early embryonic stages are quite faithfully mimicked,46 spontaneously differentiating EBs may not offer ideal conditions for the
refined processes occurring later in development. EBs lack the
anatomical structure of the embryo, and cells developing in EBs
do not participate accurately in embryonic cell-to-cell interactions,
and miss certain cell non-autonomous effects and exposure to
physical stimuli that physiologically occur at a specific stage of
development (as for example the flow pressure and cellular
movement occurring with the onset of embryonic circulation47).
It is likely that in vitro differentiating ESCs are prone to generate
immature, embryonic-type progenitor cells, which may not function properly upon transplantation in adults. However, these
apparent inconveniences at the same time harbour the chance that
– assuming specific embryonic differentiation cues are identified and provided in vitro – large homogenous populations of cells
can be generated in this system. Therefore, in vivo studies of
embryonic development in mice, or in developmental models such
as zebrafish, Xenopus and chick remain as key approaches towards
learning how to generate adult-type cells from ESC.
Another hurdle towards generation of ready-to-use adult-type
cells from human PSCs is the assay available for the evaluation of
human cells. Human cells are less characterized than murine (for
example, with respect to surface antigens or molecular signatures)
and functional evaluation is confined to surrogate xenogeneic
models which can introduce biases and not always faithfully reflect
cell function. For example, human HSCs transplanted into mice rely
on mouse cytokines and niche factors with only limited cross-reactivity to human cells,48 while human heart grafts in rodent models
are exposed to atypically high heart beat frequencies. Such environmental factors bias functional evaluation of the cells and may
contribute to reported differences between mouse and human
cells. HSCs showing long term engraftment were obtained from
murine ESCs,11 and iPS cells49 using transduction with Hoxb4 and
coculture on OP9-stromal cells, while human HSCs could not be
produced following similar protocols.50
In many transplant protocols, using iPS cells would be advantageous as the derived cells would be isogenic and would not face immune barriers in the host. The pluripotent nature of mouse iPS cells51
has been demonstrated by blastocyst chimaerization and tetraploid
complementation assays, and the proof of principle experiment that
such cells can be used for providing isogenic, genetically corrected
cellular products has been performed in a sickle-cell anemia mouse
model.49 Undifferentiated human iPS cells show a molecular signature highly similar to human ESCs,20–22 and hESC-protocols can be
used to drive their in vitro differentiation into derivatives of the three
germ layers.22 However, critical issues such as viral integration and
residual levels of transgene expression52 may impact the differentiation potential of iPS cells. Obviously, individual reprogramming
protocols can introduce distinct biases (e.g. through use of different
embryonic proteins, higher transgene persistence after use of lentiviral versus retroviral vectors, off-target effects of small molecules).51
Detailed characterization of the ability of human iPS cells to form
specific tissues and functional characterization of the resulting cells
is still in its infancy. However, recent studies provide encouraging
evidence that generation of cardiomyocytes,52 adipocytes53 and
hematopoietic and endothelial cells is similar between human ESCs
and human iPS cells.20,21,54 Data from our laboratory supports these
observations, showing robust blood cell formation that could be further enhanced by bone morphogenetic 4 supplementation (BMP4) in
differentiating human iPS cells generated by a separate protocol.22,142 Since, only scant data currently available on the differentiation potential of human iPS cells, in this review we will highlight
results reported on differentiating hESCs.
Hematopoietic development during embryogenesis: primitive
versus definitive blood
When attempting to differentiate pluripotent stem cells into
adult-type blood cells, we need to assess how differentiating EBs
recapitulate embryonic hematopoietic development. Hematopoietic and endothelial cells both arise from the mesodermal germ
layer. Common precursors called ‘‘hemangioblasts” have been
found in early stage mouse conceptuses55 and clonally identified
in in vitro differentiating mouse and human ESCs.56–58 Moreover,
vertebrate hematopoiesis occurs in two successive waves, primitive and definitive, that differ anatomically and in cell types produced. In the mouse, primitive (embryonic) blood develops
transiently in the extraembryonic yolk sac, giving rise to the first
blood cells consisting mainly of nucleated erythrocytes and macrophages. The definitive blood programme occurs subsequently at
intraembryonic sites, and lasts the life of the organism, producing
hematopoietic stem cells (HSCs) capable of extensive self-renewal
and multilineage differentiation into all blood lineages.44,59,60 Recent studies suggest that hematopoietic cells arise from hemangioblasts through a hemogenic endothelial intermediate, and show by
time-lapse microscopy hemogenic endothelium from in vitro differentiating mouse ESCs as well as early mouse embryos.61,62 It is
unclear at this point whether endothelial cells giving rise to primitive blood cells differ from those producing definitive blood cells.
Whether definitive HSCs arise in the yolk sac has been debated
for decades. The first experiments aiming to trace the origin of
blood cells were performed by Moore and Owen: chick embryo
where grafted onto a donor yolk sac, and after several days of incubation, donor yolk sac derived hematopoietic cells were found in
the hematopoietic organs of the recipient embryo, demonstrating
the ability of yolk sac cells to participate in definitive hematopoiesis.63,64 However, these experiments were performed after the onset of circulation, and contamination from circulating HSCs was
possible: with the onset of blood flow, cells from intraembryonic
sites migrate back to the yolk sac, which then might contain definitive HSCs. Repetition of the avian grafting experiments using precirculation yolk sacs detected no contribution to host embryo
hematopoiesis, arguing that indeed adult HSCs arise solely from
an intraembryonic location.65–67 Analyses indentified intraembryonic blood sites within the chick aortic endothelium68 and cells
displaying hallmark properties of adult HSCs (longterm repopulation and multilineage differentiation upon transplantation in irradiated adult hosts) in the murine aorto-gonado-mesonephros
region (AGM).69,70 However, cells from precirculation murine yolk
sacs can also give rise to definitive HSCs, if they are transplanted
into the supportive environment of the fetal liver of newborn
mice.71,72
Generating blood from PSCs
Analysis of in vitro differentiating PSCs reveal a close resemblance to in vivo embryonic processes:46,73,74 supplementation of
PSCs cultures with morphogens and inductive signals known from
in vivo developmental models (e.g. zebrafish, frog, and chick) enables directed differentiation in the absence of serum-containing
medium46 (reviewed in44). When directed by morphogens like
bone morphogenetic protein (BMP), Wingless (Wnt) and Activin,
ESCs develop into cells equivalent to the primitive streak, and give
rise to cells of the three germ layers (ectoderm, endoderm and
mesoderm).45 Gene expression analysis distinguishes between
the formation of anterior primitive streak-like cells, developing
mostly into endoderm, and posterior primitive streak-like cells
that will differentiate into mesoderm and from there through activation of patterning genes (e.g. Cdx/Hox), into hemangioblasts45
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
and blood cells. Mouse hemangioblasts express the mesodermal
marker Brachyury and can be identified around day 3 in differentiating EBs by expression of fetal liver kinase 1 (Flk1).58 Human
hemangioblasts56 are phenotypically less well described, yet are
found in the CD34+CD45 cellular population75 and express KDR
(VEGF receptor 2),56 CD31,76 and angiotensin-converting-enzyme
(ACE/CD143).77 With maturation along the blood lineage, these
progenitors start expressing the hematopoietic antigen CD45.78
Emergence of CD34+CD45+ cells correlates with derivation of
hematopoietic progenitor cells with colony forming unit (CFU) potential in methylcellulose assays, and sorting for CD34+CD45+ cells
enriches for cells giving rise to hematopoietic colonies.76
Two different protocols, or combinations of them, can be used
for generating hematopoietic progenitors from PSCs: (1) formation
of embryoid bodies (EBs) and (2) culture on supportive stromal
layers79 (e.g. OP9-stromal cells, reviewed in 44). What type of
hematopoietic progenitors is routinely generated from ESCs
remains as an intriguing question. Most probably, ESCs readily give
rise to yolk-sac-type progenitors, generating primitive erythroid
cells as well as macrophages, definitive erythroid cells, megakaryocytes and mast cell lineages, and do not routinely differentiate
into specific lymphoid cells or true hematopoietic stem cells
(HSC). However, if exposed to specific conditions, ESCs do form
B-80 and T-81 lymphoid cells. Thus, even though yolk-sac-type
hematopoiesis is predominant, definitive hematopoietic cells may
be obtained from in vitro differentiating ESCs.
PSCs-derived HSCs
The ultimate goal remains generating ‘‘off-the-shelf” PSC-derived human HSCs that will be capable of repopulation and durable
reconstitution of the entire human hematopoietic system in adults.
There are several reports on murine ESC-derived hematopoietic
elements capable of long term multilineage engraftment (reviewed
in,44 Fig. 1). Burt and colleagues reported the formation of ckit+CD45+ transplantable hematopoietic progenitors, capable of
long term multilineage reconstitution of mice, following culture
of mESCs in methylcellulose in the presence of serum, stem cell
factor, IL-3 and IL-6.82 Importantly, direct delivery to the bone
marrow via intrafemoral instillation enabled significantly higher
numbers of engrafted cells, as compared to intravenous application
in the tail vein.82 These findings are consistent with results from
other studies documenting the superiority of intra-bone marrow
over intravenous transplantation,83,84 and suggest that providing
direct contact with the niche may be especially important for
31
developmentally immature stem cells. Interestingly, applying high
numbers of purified c-kit+CD45+ cells enabled engraftment even in
MHC-mismatched mice, without signs of graft rejection of induction of GvHD. Tolerance induction through high numbers of transplanted material may be another advantage offered by the PSCsystem. However, the encouraging results reported in this study
have not been yet independently replicated, as they also have
not for the early study of Palacios and colleagues who reported
back in 1995 robust multilineage repopulation following coculture
with the stromal cell line RP010.85 In this latter case, replication of
results has been hampered by the fact that RP010 cells are not
readily available.
Coculture with stromal cell lines (e.g. derived from AGM or fetal
liver, where HSCs form and expand during their in vivo embryonic
development) is an appealing way of promoting maturation and
expansion of PSC-derived blood progenitors. A prominent example
is the well-known coculture with OP9-stromal cells, which provides a supportive microenvironment,50 generating enhanced
hematopoietic activity and promoting lymphogenesis. OP9-stromal cells are derived from the calvariae of newborn op/op mice
which lack macrophage colony-stimulating factor (M-CSF).86 The
absence of M-CSF inhibits the survival of monocyte-macrophage
cells, which otherwise overwhelm other lineages. If, in addition
to coculture on OP9-stromal cell, enforced expression of the patterning genes Cdx4/HoxB4 is performed, it is possible to generate
true ESC-derived murine HSCs, capable of robust multilineage
hematopoietic reconstitution in irradiated adult mice.11,50,87 Several groups have reported production of repopulating HSCs following genetic modification of mESC-progenitors with the homeobox
gene HoxB4 and expansion on OP9-stroma cells.11,24,88 Following
this protocol, repopulating murine HSCs have been efficiently generated also from pluripotent stem cells of different origin (e.g. parthenogenetic89 and reprogrammed iPS cells,49 Table 1). Homeobox
(Hox) genes are transcription factors involved in embryonic tissue
patterning processes, and play important roles in developmental
hematopoiesis as well as homeostasis and malignant transformation of adult blood cells (reviewed in90). Particularly HoxB4 has
been shown to enhance self-renewal and/or growth activity of
hematopoietic progenitors and HSCs in mouse and human
cells.91–93 In the immature murine yolk sac or ESC-derived hematopoietic progenitors, enforced HoxB4 expression renders competence for long term multilineage engraftment of irradiated adult
hosts.11 However, for reasons that are not clear, lymphoid engraftment appears less robust than myeloid engraftment in these cells.
Possibly, persisting HoxB4 overexpression biases further differentiation potential of the transplanted cells away from the lymphoid
Fig. 1. Blood cells from pluripotent stem cells. Mouse ES cells expressing ectopic HoxB4 differentiate on OP9 stroma into repopulating HSC. (A) Hematopoietic cells from day 6
embryoid body-derived cells after seven days of co-culture with OP9 stroma. Human iPS cells readily give rise to multiple blood cells including erythroid progenitors, yet have
not yet been efficiently differentiated into HSC. (B) BFU-E colony emerging from embryoid body-derived cells plated in methylcellulose supplemented with hematopoietic
cytokines. Abbreviations: BFU-E (burst forming colony-erythroid); ES (embryonic stem); HSC (hematopoietic stem cells); iPS (induced pluripotent stem); OP9 (murine MCSF-/- stromal line).
32
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
lineage.88 Engraftment rates, especially also engraftment with lymphoid cells, can be enhanced by combinatorial transduction with
Cdx4, a homeobox transcription factor of the caudal homeobox
gene family. cdx genes have been first discovered as early patterning regulators of hematopoietic fate in zebrafish and later shown to
promote hematopoiesis from murine ESCs through regulation of
downstream posterior Hox genes.74,87,94–97 Moreover, recent studies suggest that CDX genes involvement in human leukemogenesis
of the myeloid as well of the lymphoid lineage,98–101 reinforcing
the notion that molecular pathways are shared between developmental and adult hematopoiesis (as seen also with Hox genes).
However, this approach requires genetic modification and has
not been successful with human cells: cocultured with OP9 and
transduction with HoxB4 cells promotes hematopoietic activity
from human ESCs, by inhibiting apoptosis of ESCs-derived
CD34+CD45+ cells; however, it does not confer stem cell function
to human progenitors (see below).50 Recently, Ledran and colleagues reported that the culture of human ESCs on a number of
stromal cell lines and primary cells derived from the AGM and fetal
liver significantly enhanced hematopoietic activity, including
hematopoietic engraftment capacity into immunocompromised
mice in primary and secondary transplant assays.79 However, further studies are needed to confirm these data and to improve
chimaerism.
While transduction with HoxB4 and/or Cdx4 promotes robust
engraftment, cells exhibiting the classical surface antigen phenotype of engraftable murine HSC (c-kit+Sca1+Lin ) have not yet been
reported in this system. To our knowledge, CD41 is the earliest
antigen characterizing preformed embryonic blood progenitors,
as assayed by in vitro colony forming assays and gene expression
data96,102 from murine ESCs and mouse embryos. In day 6 EB,
hematopoietic activity is confined to cells expressing CD41+, which
represent approximately 30% of EB-derived under differentiation in
serum-containing medium. Within the CD41+ compartment, colony forming activity is highly enriched among CD41+c-kit+ double
positive cells, suggesting that c-kit marks cells with stem/progenitor capacity and may be downregulated with differentiation
(CD41+ckit cells). Only CD41+(c-kit+) cells are able to form colonies on OP9-stroma cells, which is a necessary step in the development of transplantable cells. Ectopic expression of Cdx4 enhances
the hematopoietic activity of CD41+ckit+ cells, without conferring
hematopoietic potential to CD41 cells.96 The panhematopoietic
antigen CD45 appears shortly after CD41 (day 6.5–7 in differentiating EB), presumably indicating conversion of CD41+ cells to more
mature progenitors. However, colony forming potential declines
with the appearance of CD45, suggesting that further development
in EB rapidly promotes terminal differentiation of the CD41+ progenitor cells (Lengerke C, unpublished observation). Recent comparison of the phenotype of ESC-derived repopulating HSCs with
hematopoietic stem and progenitor cells derived from distinct
in vivo developmental stages (murine yolk sac, aorta-gonad-mesonephros, placenta, fetal liver and bone marrow) suggests that ESCderived HSCs are a developmentally immature population of cells
with features of both primitive and mature HSCs, defined as
ckit+CD41+CD34 CD150+CD45+/ CD48+/ ).103 Further studies are
needed to characterize these cells from human ESCs.
Generating human HSCs from hESCs remains challenging
(Table 3). As in the murine system, human ESCs differentiate robustly
into the hematopoietic lineage in EBs, as well as in coculture systems with supportive stromal cells. Cells with features resembling
adult-type HSCs are produced: e.g. CD34+ expression, the ability to
efflux Hoechst dye, high aldehyde dehydrogenase activity and multilineage hematopoietic colony potential in clonal assays.104 However, very limited repopulation ability is observed in xenogeneic
transplant assays.105 HoxB4 enhances hematopoietic activity of human blood progenitor cells,93 and OP9-stromal cell cocultures aug-
ment survival of hESC-derived CD34+CD45+ hematopoietic
precursors. However, neither HoxB4 transduction nor OP9-stroma
coculture were able to confer HSC activity to hESC-derived
cells.50,105 Interestingly, OP9-stroma coculture effects the
CD45+CD34+, but not the more differentiated CD45+CD34 cellular
compartment, indicating specific effects on hematopoietic progenitor cells.50 The fact that HoxB4 does not promote HSC formation
from hESCs could be the result of technical issues or could reflect
intrinsic differences in the biology of human and mouse ESC-derived hematopoietic cells. While differentiation in the murine system proves that derivation of HSCs is possible, strategies for
human ESCs still need to be developed. To facilitate transition to
clinical protocols, approaches involving animal products and
coculture systems should be replaced and stable transduction
through inserting viral vectors avoided, replaced by strategies
involving adenoviral gene transfer, protein transduction,106 or
small molecules.
Production of specific blood lineages from PSC
While HSCs remain difficult to obtain, there are several reports
of successful derivation of specific lineages from ESCs by supplementing cultures with cytokines or chemicals or/and using coculture with specific stroma cell lines. Robust generation of murine
erythroid cells and self-renewing immature erythroid progenitors,107 megakaryocytes,108,109 granulocytes,110,111 mast cells,112
eosinophils,113 T and B lymphocytes,81,86,114–116 macrophages,117
dendritic cells,118–120 NK cells121 has been reported from mESCs
(reviewed by Olsen and colleagues122). This work in mESCs has laid
the foundation for interrogating detailed aspects of blood cell biology, such as the role of GATA1 during proerythroblast maturation,123 and provided a basis for the development of similar
protocols in hESC,122 opening up prospects for use in cell replacement therapies. We will review here recent progress that has been
made in blood cell production from hESCs (Table 2).
Red blood cells from human PSC
Large-scale production of red blood cells from the human ESC
line H1 was reported 2006 by Olivier et al. High numbers of erythrocytes (5000-fold increase in cell number) were obtained by a 14day coculture with immortalized human fetal liver cells in the
presence of serum-containing medium and subsequent expansion
of CD34+ cells in serum-free liquid cultures supplemented with
cytokines and defined factors (hydrocortisone, IL3, BMP4, Flt-3L,
SCF, EPO, IGF-1, and hemin).124 However, the red blood cells obtained were mostly nucleated primitive erythroblasts, which expressed a mixture of embryonic and fetal globins but not betaglobin characteristic for adult cells.124 Two years later, the same
laboratory reported that prolonging the differentiation cultures
on fetal liver cells to 35 days promotes the differentiation of fetal
liver-like erythroblasts which are smaller in size, express mostly
fetal hemoglobin and are able to enucleate, suggesting that hESCderived erythropoiesis mimics human development and recapitulates accordingly the globin gene switch.125 Functional exploration
of hESC-derived red blood cells showed oxygen equilibrium curves
comparable to normal red blood cells and, adequate responses to
pH and 2,3-phoshodiglycerate changes and suggests further
in vitro maturation to adult-type beta-globin expressing
cells.126,127 One study reports differentiation to functional erythrocytes under serum-free conditions, by using supplementation with
cell-permeable recombinant HoxB4 transcription factor.126 While
these data provide encouraging results and a highly valuable model system to study genetic regulation of erythropoiesis, several issues need to be resolved before manufacture of red blood cells can
become a clinical application:128 (1) the derivation methods need
33
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
Table 2
Blood cell types generated by in vitro differentiation of human embryonic stem cells.
Blood lineage
hESC lines
Differentiation protocol
Cytokines and growth factors
Key
references
HSC
Primitive erythroid
–
HI
–
Hydrocortisone IL-3, BMP4, SCF, Flt-3
ligand, EPO, IGF-1, hemin
–
124
Definitive erythroid
HI
Hl, HUES-3 MA01, MA99
–
14 Days coculture on immortalized fetal
liver cells
Expansion in serum-free liquid cultures
Longer periods of culture (>35 days)
Recombinant tPTD-HoxB4 fusion protein
125
Megakaryocytes and
platelets
KhES-1,-2,-3
H9, HSF6
Neutrophils
KhES-3
VEGF-induced ESC-derived sacs and coculture with OP9 or C3H10T1/2 stroma
Coculture with OP9-stroma cells
Coculture with OP9-stroma cells
See above
BMP4, VEGF, bFGF, SCF, TPO, Flt-3
ligand, EPO
VEGF, TPO
TPO
132
Monocytes and
Macrophages
NK cells
KCL001, KCL002, HUES-2
EB formation, no cocultures
BMP4, SCF, Flt-3 ligand, IL-3, IL-6, FP6,
TPO, G-SCF
M-CSF, IL-3
H9
Cocultures with the bone marrow stromal
cell line M210-B4136 or the stromal cell line
S1713 followed by cocultures with the murine fetal liver line AFT024
Differentiation on OP9-stroma138 or in
EB,139 followed by injection into human
thymic tissue implanted in immunocompromised mice
Coculture on Deltal-expressing OP9-stromal cells
Predifferentiation in coculture with OP9,
lymphomyeloid differentiation on MS-5
stroma
IL-3, IL-15, IL-7, SCF, Flt-3 ligand136
136,137
BMP4, SCF, Flt-3 ligand
SCF, IL-7, Flt-3 ligand
138,139
140
SCF, Flt-3 ligand, IL-3, IL-7, TPO, VEGF,
BMP4
104
T-lymphocytes
HI
HI
B-lymphocytes
H1, H9
131
134
133
Abbreviations: bFGF, basic fibroblast growth factor; BMP, bone morphogenetic protein; hESC, human embryonic stem cells; IL, interleukin; EPO, erythropoietin; Flt-3 ligand,
fms tyrosine like 3 ligand; G-CSF, granulocyte colony-stimulating factor; M-CSF, macrophage colony-stimulating factor; OP9, murine M-CSF-/- stromal line; NK, natural killer;
SCF, stem cell factor; TPO, thrombopoietin; VEGF, vascular endothelial growth factor.
to be performed serum-free; (2) in a highly scalable fashion (up to
now, the largest number reported was 2 billion red blood cells; in
comparison, one 220-ml unit of packed red blood cells contains
about 2 trillion cells); (3) at reasonable cost enabling industrial
production; (4) with improved generation of adult-type beta
hemoglobin expressing cells. Questions regarding half-life, immunogenicity, contamination with potentially tumorigenic cells need
to be addressed before ESC-derived red blood cells can enter largescale clinical applications and replace conventional red blood cell
supplies.
While the issues listed above may hamper applications on a
large industrial scale, there may be individual benefit for patients
particularly in need of customized blood cells, for example patients
requiring lifelong erythrocyte substitution therapies that over time
develop alloantibodies that highly restrict the pool of matching
blood cell donors. It is plausible that generating patient-specific
blood cells from their own pluripotent stem cells will benefit and
increase treatment possibilities in these patients.
Granulocytes and platelets from human PSCs
Granulocytes are not routinely provided by the current donation-based blood transfusion system. PSCs may represent a source
for generation of large amounts of neutrophils for the treatment of
patients presenting with life-threatening neutropenia. Several
studies report efficient formation of myeloid cells from mouse
and human ESCs using combinatorial differentiation approaches
in EBs and coculture strategies with stromal cell lines (e.g. S17129
and OP9104).78,104,105,129–132 In a recent report, Yokoyama and colleagues provide evidence that functional neutrophils can be produced from hESCs by using EB-differentiation in serum-based
medium followed by coculture on OP9 cells for 7–14 days in medium supplemented with BMP4, SCF, Flt-3L, IL6, FP6, TPO and G-SCF.
During culture on OP9, hESC-derived progenitors matured from
mostly myeloblasts and promyelocytes on day 7 to matured termi-
nally differentiated neutrophils on day 14. The vast majority of
OP9-cocultured progenitors expressed the hematopoietic marker
CD45 at all timepoints, while expression of primitive cell markers
(CD133, CD34 and CD117) was lost at later timepoints, indicating
maturation to terminally differentiated neutrophils. Moreover,
functional comparison of hESC- and human peripheral blood derived neutrophils showed similar phagocytosis, chemotaxis, superoxide production, bactericidal activity and oxidative burst capacity,
despite observed slight differences in cellular phenotype (hESC-derived neutrophils show decreased CD16 expression and aberrant
CD64 and CD14 expression).132 Using similar approaches but different combinations of growth factors and cytokines (M-CSF, IL3), other reports show homogeneous production of functional
monocytes and macrophages from hESCs, providing tools for investigating myeloid cell development and biology.133 Megakaryocyte
formation has also been reported from human hESCs.131,134 Using
differentiation on OP9-stromal cells and supplementation with
VEGF and TPO, Takayama and colleagues report generation of multipotent hematopoietic progenitors and efficient production of mature megakaryocytes expressing specific surface antigens such as
CD41a, CD42a and CD42b.131 Some megakaryocytes appeared to
be shedding their cytoplasmic membranes, displaying demarcation
membrane systems necessary for platelet formation, and plateletlike particles were detected in culture supernatants. Electron
microscopy confirmed normal microtubule formation, similar to
plasma-derived human platelets, although ESC-derived platelets
displayed fewer granules.131 Stimulation of hESC-derived platelets
with thrombin and ADP induced GPIIb/IIIa activation, and filopodia
formed upon adherence to fibrogen-coated dishes, indicating
functionality of hESC-derived platelets. On average, after 24 days
of culture, approximately 5 106 platelets were generated from
an initial 105 human ESCs. hESCs megakaryocytes yield fewer platelets in vitro than their adult counterparts in vivo, possibly due to
some microenvironment stimulus lacking in the in vitro differentiation system (e.g. shear flow135). Establishing in vitro protocols for
34
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
Table 3
Human blood cell therapies from pluripotent stem cells: limitations and some potential solutions.
Limitations
Possible solutions
Generation of mostly primitive blood progenitors
instead of definitive type-HSC
Improving HSC generation by optimizing the in vitro differentiation protocols using:
– morphogens,46 cytokines, small molecules and
– biomechanical stimuli mimicking the in vivo developmental environment47
Development of non-HSC blood cell therapies:
– erythroid progenitors
– megakaryocytes
– neutrophils
– NK cells (e.g. engineered with anti-tumor activity)
HSC identification
HSC maintenance
Tumorigenicity and safety
Homing deficiencies
Assessment of HSC/progenitors in
xenotransplantation assays
Characterization of the fetal HSC phenotype
Discovery of protocols based on cytokines and small molecules
Transplantation of well differentiated (sorted) progenitors
Engineering of inducible suicide programs
Transplantation directly into the bone marrow
Supplementation of non-/(weakly)-cross reactive mouse cytokines with human counterparts creation of a
human stem cell niche by cotransplantation of human stromal cells
efficient human platelet generation from human PSCs promises
the production of isogenic or histocompatible platelets that can
potentially circumvent the need to obtain platelets through blood
donation. One advantage in this system is that, as with erythrocytes, mature platelets are enucleated cells that can be irradiated,
thereby eliminating safety concerns due to contamination with
residual undifferentiated cells.
Lymphoid cell formation from human PSCs
While erythroid and myeloid progenitor cells can be routinely
generated from human PSCs, there are few reports on the generation of lymphoid cells in this system. In a recent report, Woll and
colleagues demonstrated generation of functional hESC-derived
NK cells capable of inducing efficient anti-tumor responses
in vivo. hESC-derived NK cells are uniformly CD94+CD117low/and present higher tumor-clearing capacity than NK cells derived
from umbilical cord blood,136 suggesting that they may serve as a
novel cellular source for anti-tumor immunotherapy. Although NK
and T cells are developmentally closely related, hESC-derived
hematopoietic progenitors develop into natural killer (NK)
cells,136,137 but typically do not form T- or B-cells,137 unless specific
conditions are provided (e.g. T-cell development following injection into human thymus/fetal liver grafts in severe combined
immunodeficient-humanized (SCID-hu) mice138,139 or coculture
conditions with OP9-stromal cells expressing high Delta-1 like
activity140). Under such conditions, robust T-cell development
was observed from hESCs,138 even though in a less efficient manner
than from human fetal liver, bone marrow or umbilical cord hematopoietic progenitors. Another study was able to show under OP9coculture conditions small percentages of CD19+ cells from hESCs,
indicating their potential to form B-cells.104 Taken together, these
studies corroborate the hypothesis that ESC differentiation protocols drive embryonic hematopoiesis and formation of progenitor
cells resembling human yolk sac derived CD34+ cells,47,141 but
identifying specific environmental cues will be essential to induce
formation of adult-type cells.
Conclusion
ESCs and the more recently developed iPS cells hold promise for
generating histocompatible or isogenic cellular transplant therapies for a variety of patients. Significant hurdles on the way to clinical application are the biased differentiation into rather immature,
embryonic-like progenitor cells that may not functional normally
in adults. Furthermore, differentiation protocols for clinical stan-
dards need to be improved by removing serum, and other such animal products, as well as cocultures with animal cells. However,
PSCs offer an exciting new model enabling unique studies on human hematopoietic development and disease (reviewed).143
Conflict of interests statement
George Daley is a member of the Scientific Advisory Board of
IPierian (iPS cells for drug discovery) and Epizyme (cancer drugs
targeting chromatin modifying enzymes).
Acknowledgements
C.L. is supported by grants from the DFG SFB773, the Deutsche
Krebshilfe Max-Eder-Program and the Fortune Program of the University of Tuebingen. G.Q.D. is a recipient of the NIH Director’s Pioneer Award of the NIH Roadmap for Medical Research, Clinical
Scientist Awards in Translational Research from the Burroughs
Wellcome Fund and the Leukemia and Lymphoma Society and supported by grants from the United States National Institutes of
Health and the Howard Hughes Medical Institute. We thank Shannon McKinney-Freeman and Odelya Hartung for providing the
photos of pluripotent stem cells-derived hematopoietic cells.
References
1. Martin GR. Isolation of a pluripotent cell line from early mouse embryos
cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad
Sci USA 1981;78(12):7634–8.
2. Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from
mouse embryos. Nature 1981;292(5819):154–6.
3. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines
derived from human blastocysts. Science 1998;282(5391):1145–7.
4. Thomas ED, Lochte Jr HL, Lu WC, Ferrebee JW. Intravenous infusion of bone
marrow in patients receiving radiation and chemotherapy. N Engl J Med
1957;257(11):491–6.
5. Park IH, Arora N, Huo H, et al. Disease-specific induced pluripotent stem cells.
Cell 2008;134(5):877–86.
6. Hacein-Bey-Abina S, Le Deist F, Carlier F, et al. Sustained correction of X-linked
severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med
2002;346(16):1185–93.
7. Drukker M, Katz G, Urbach A, et al. Characterization of the expression of MHC
proteins in human embryonic stem cells. Proc Natl Acad Sci USA
2002;99(15):9864–9.
8. Briggs R, King TJ. Transplantation of living nuclei from blastula cells into
enucleated frogs’ eggs. Proc Natl Acad Sci USA 1952;38(5):455–63.
9. Gurdon JB. The developmental capacity of nuclei taken from intestinal
epithelium cells of feeding tadpoles. J Embryol Exp Morphol 1962;10:622–40.
10. Gurdon JB. Adult frogs derived from the nuclei of single somatic cells. Dev Biol
1962;4:256–73.
11. Kyba M, Perlingeiro RC, Daley GQ. HoxB4 confers definitive lymphoid-myeloid
engraftment potential on embryonic stem cell and yolk sac hematopoietic
progenitors. Cell 2002;109(1):29–37.
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
12. Rideout 3rd WM, Hochedlinger K, Kyba M, Daley GQ, Jaenisch R. Correction of
a genetic defect by nuclear transplantation and combined cell and gene
therapy. Cell 2002;109(1):17–27.
13. Barberi T, Klivenyi P, Calingasan NY, et al. Neural subtype specification of
fertilization and nuclear transfer embryonic stem cells and application in
parkinsonian mice. Nat Biotechnol 2003;21(10):1200–7.
14. Stojkovic M, Stojkovic P, Leary C, et al. Derivation of a human blastocyst after
heterologous nuclear transfer to donated oocytes. Reprod Biomed Online
2005;11(2):226–31.
15. Muller R, Lengerke C. Patient-specific pluripotent stem cells: promises and
challenges. Nat Rev Endocrinol 2009;5(4):195–203.
16. Kim K, Lerou P, Yabuuchi A, et al. Histocompatible embryonic stem cells by
parthenogenesis. Science 2007;315(5811):482–6.
17. Revazova ES, Turovets NA, Kochetkova OD, et al. Patient-specific stem cell
lines derived from human parthenogenetic blastocysts. Cloning Stem Cells
2007;9(3):432–49.
18. Revazova ES, Turovets NA, Kochetkova OD, et al. HLA homozygous stem cell
lines derived from human parthenogenetic blastocysts. Cloning Stem Cells
2008;10(1):11–24.
19. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse
embryonic and adult fibroblast cultures by defined factors. Cell
2006;126(4):663–76.
20. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells
from adult human fibroblasts by defined factors. Cell 2007;131(5):861–72.
21. Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines
derived from human somatic cells. Science 2007;318(5858):1917–20.
22. Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to
pluripotency with defined factors. Nature 2008;451(7175):141–6.
23. Aoi T, Yae K, Nakagawa M, et al. Generation of pluripotent stem cells from
adult mouse liver and stomach cells. Science 2008;321(5889):699–702.
24. Kim JB, Zaehres H, Wu G, et al. Pluripotent stem cells induced from adult
neural stem cells by reprogramming with two factors. Nature
2008;454(7204):646–50.
25. Kim JB, Sebastiano V, Wu G, et al. Oct4-induced pluripotency in adult neural
stem cells. Cell 2009;136(3):411–9.
26. Stadtfeld M, Brennand K, Hochedlinger K. Reprogramming of pancreatic beta
cells into induced pluripotent stem cells. Curr Biol 2008;18(12):890–4.
27. Hanna J, Markoulaki S, Schorderet P, et al. Direct reprogramming of terminally
differentiated
mature
B
lymphocytes
to
pluripotency.
Cell
2008;133(2):250–64.
28. Loh YH, Agarwal S, Park IH, et al. Generation of induced pluripotent stem cells
from human blood. Blood.
29. Huangfu D, Osafune K, Maehr R, et al. Induction of pluripotent stem cells from
primary human fibroblasts with only Oct4 and Sox2. Nat Biotechnol.
30. Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by
defined factors is greatly improved by small-molecule compounds. Nat
Biotechnol 2008;26(7):795–7.
31. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent
stem cells generated without viral integration. Science.
32. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of
mouse induced pluripotent stem cells without viral vectors. Science
2008;322(5903):949–53.
33. Woltjen K, Michael IP, Mohseni P, et al. PiggyBac transposition reprograms
fibroblasts to induced pluripotent stem cells. Nature 2009;458(7239):766–70.
34. Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free
induction of pluripotency and subsequent excision of reprogramming factors.
Nature 2009;458(7239):771–5.
35. Matsui Y, Zsebo K, Hogan BL. Derivation of pluripotential embryonic stem cells
from murine primordial germ cells in culture. Cell 1992;70(5):841–7.
36. Shamblott MJ, Axelman J, Wang S, et al. Derivation of pluripotent stem cells
from cultured human primordial germ cells. Proc Natl Acad Sci USA
1998;95(23):13726–31.
37. Guan K, Nayernia K, Maier LS, et al. Pluripotency of spermatogonial stem cells
from adult mouse testis. Nature 2006;440(7088):1199–203.
38. Kanatsu-Shinohara M, Inoue K, Lee J, et al. Generation of pluripotent stem cells
from neonatal mouse testis. Cell 2004;119(7):1001–12.
39. Seandel M, James D, Shmelkov SV, et al. Generation of functional multipotent
adult stem cells from GPR125+ germline progenitors. Nature
2007;449(7160):346–50.
40. Conrad S, Renninger M, Hennenlotter J, et al. Generation of pluripotent stem
cells from adult human testis. Nature.
41. Ohtaka T, Matsui Y, Obinata M. Hematopoietic development of primordial
germ cell-derived mouse embryonic germ cells in culture. Biochem Biophys Res
Commun 1999;260(2):475–82.
42. Doetschman TC, Eistetter H, Katz M, Schmidt W, Kemler R. The in vitro
development of blastocyst-derived embryonic stem cell lines: formation of
visceral yolk sac, blood islands and myocardium. J Embryol Exp Morphol
1985;87:27–45.
43. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically
relevant populations: lessons from embryonic development. Cell
2008;132(4):661–80.
44. Lengerke C, Daley GQ. Patterning definitive hematopoietic stem cells from
embryonic stem cells. Exp Hematol 2005;33(9):971–9.
45. Gadue P, Huber TL, Paddison PJ, Keller GM. Wnt and TGF-beta signaling are
required for the induction of an in vitro model of primitive streak formation
using embryonic stem cells. Proc Natl Acad Sci USA 2006;103(45):16806–11.
35
46. Lengerke C, Schmitt S, Bowman TV, et al. BMP and Wnt specify
hematopoietic fate by activation of the Cdx-Hox pathway. Cell Stem Cell
2008;2(1):72–82.
47. Adamo L, Naveiras O, Wenzel PL, et al. Biomechanical forces promote
embryonic haematopoiesis. Nature 2009;459(7250):1131–5.
48. Manz MG. Human-hemato-lymphoid-system mice: opportunities and
challenges. Immunity 2007;26(5):537–41.
49. Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse
model with iPS cells generated from autologous skin. Science
2007;318(5858):1920–3.
50. Ji J, Vijayaragavan K, Bosse M, Menendez P, Weisel K, Bhatia M. OP9 stroma
augments survival of hematopoietic precursors and progenitors during
hematopoietic differentiation from human embryonic stem cells. Stem cells
2008;26(10):2485–95.
51. Maherali N, Sridharan R, Xie W, et al. Directly reprogrammed fibroblasts show
global epigenetic remodeling and widespread tissue contribution. Cell Stem
Cell 2007;1(1):55–70.
52. Zhang J, Wilson GF, Soerens AG, et al. Functional cardiomyocytes derived from
human induced pluripotent stem cells. Circ Res 2009;104(4):e30–41.
53. Taura D, Noguchi M, Sone M, et al. Adipogenic differentiation of human
induced pluripotent stem cells: comparison with that of human embryonic
stem cells. FEBS Lett 2009;583(6):1029–33.
54. Choi KD, Yu J, Smuga-Otto K, et al. Hematopoietic and endothelial
differentiation of human induced pluripotent stem cells. Stem Cells
2009;27(3):559–67.
55. Huber TL, Kouskoff V, Fehling HJ, Palis J, Keller G. Haemangioblast
commitment is initiated in the primitive streak of the mouse embryo.
Nature 2004;432(7017):625–30.
56. Kennedy M, D’Souza SL, Lynch-Kattman M, Schwantz S, Keller G. Development
of the hemangioblast defines the onset of hematopoiesis in human ES cell
differentiation cultures. Blood 2007;109(7):2679–87.
57. Kennedy M, Firpo M, Choi K, et al. A common precursor for primitive
erythropoiesis
and
definitive
haematopoiesis.
Nature
1997;386(6624):488–93.
58. Choi K, Kennedy M, Kazarov A, Papadimitriou JC, Keller G. A common
precursor for hematopoietic and endothelial cells. Development
1998;125(4):725–32.
59. Galloway JL, Zon LI. Ontogeny of hematopoiesis: examining the emergence of
hematopoietic cells in the vertebrate embryo. Curr Top Dev Biol
2003;53:139–58.
60. Dzierzak E, Speck NA. Of lineage and legacy: the development of mammalian
hematopoietic stem cells. Nat Immunol 2008;9(2):129–36.
61. Lancrin C, Sroczynska P, Stephenson C, Allen T, Kouskoff V, Lacaud G. The
haemangioblast generates haematopoietic cells through a haemogenic
endothelium stage. Nature 2009;457(7231):892–5.
62. Eilken HM, Nishikawa S, Schroeder T. Continuous single-cell imaging of blood
generation from haemogenic endothelium. Nature 2009;457(7231):896–900.
63. Moore MA, Owen JJ. Chromosome marker studies on the development of the
haemopoietic system in the chick embryo. Nature 1965;208(5014):956
passim.
64. Moore MA, Owen JJ. Chromosome marker studies in the irradiated chick
embryo. Nature 1967;215(5105):1081–2.
65. Dieterlen-Lievre F. On the origin of haemopoietic stem cells in the avian
embryo:
an
experimental
approach.
J
Embryol
Exp
Morphol
1975;33(3):607–19.
66. Lassila O, Eskola J, Toivanen P, Martin C, Dieterlen-Lievre F. The origin of
lymphoid stem cells studied in chick yolk sac-embryo chimaeras. Nature
1978;272(5651):353–4.
67. Lassila O, Martin C, Toivanen P, Dieterlen-Lievre F. Erythropoiesis and
lymphopoiesis in the chick yolk-sac-embryo chimeras: contribution of yolk
sac and intraembryonic stem cells. Blood 1982;59(2):377–81.
68. Dieterlen-Lievre F, Martin C. Diffuse intraembryonic hemopoiesis in normal
and chimeric avian development. Dev Biol 1981;88(1):180–91.
69. Medvinsky AL, Samoylina NL, Muller AM, Dzierzak EA. An early pre-liver
intraembryonic source of CFU-S in the developing mouse. Nature
1993;364(6432):64–7.
70. Muller AM, Medvinsky A, Strouboulis J, Grosveld F, Dzierzak E. Development
of hematopoietic stem cell activity in the mouse embryo. Immunity
1994;1(4):291–301.
71. Yoder MC, Hiatt K, Mukherjee P. In vivo repopulating hematopoietic stem cells
are present in the murine yolk sac at day 9.0 postcoitus. Proc Natl Acad Sci USA
1997;94(13):6776–80.
72. Yoder MC, Hiatt K. Engraftment of embryonic hematopoietic cells in
conditioned newborn recipients. Blood 1997;89(6):2176–83.
73. North TE, Goessling W, Walkley CR, et al. Prostaglandin E2 regulates
vertebrate
haematopoietic
stem
cell
homeostasis.
Nature
2007;447(7147):1007–11.
74. Davidson AJ, Ernst P, Wang Y, et al. Cdx4 mutants fail to specify blood
progenitors and can be rescued by multiple hox genes. Nature
2003;425(6955):300–6.
75. Zambidis ET, Oberlin E, Tavian M, Peault B. Blood-forming endothelium in
human ontogeny: lessons from in utero development and embryonic stem cell
culture. Trends Cardiovasc Med 2006;16(3):95–101.
76. Tian X, Woll PS, Morris JK, Linehan JL, Kaufman DS. Hematopoietic
engraftment of human embryonic stem cell-derived cells is regulated by
recipient innate immunity. Stem Cells 2006;24(5):1370–80.
36
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
77. Zambidis ET, Park TS, Yu W, et al. Expression of angiotensin-converting
enzyme (CD143) identifies and regulates primitive hemangioblasts derived
from human pluripotent stem cells. Blood 2008;112(9):3601–14.
78. Chadwick K, Wang L, Li L, et al. Cytokines and BMP-4 promote hematopoietic
differentiation of human embryonic stem cells. Blood 2003;102(3):906–15.
79. Ledran MH, Krassowska A, Armstrong L, et al. Efficient hematopoietic
differentiation of human embryonic stem cells on stromal cells derived from
hematopoietic niches. Cell Stem Cell 2008;3(1):85–98.
80. Cho SK, Webber TD, Carlyle JR, Nakano T, Lewis SM, Zuniga-Pflucker JC.
Functional characterization of B lymphocytes generated in vitro from
embryonic stem cells. Proc Natl Acad Sci USA 1999;96(17):9797–802.
81. Schmitt TM, de Pooter RF, Gronski MA, Cho SK, Ohashi PS, ZunigaPflucker JC. Induction of T cell development and establishment of T cell
competence from embryonic stem cells differentiated in vitro. Nat
Immunol 2004;5(4):410–7.
82. Burt RK, Verda L, Kim DA, Oyama Y, Luo K, Link C. Embryonic stem cells as an
alternate marrow donor source: engraftment without graft-versus-host
disease. J Exp Med 2004;199(7):895–904.
83. Kushida T, Inaba M, Hisha H, et al. Intra-bone marrow injection of allogeneic
bone marrow cells: a powerful new strategy for treatment of intractable
autoimmune diseases in MRL/lpr mice. Blood 2001;97(10):3292–9.
84. Ikehara S. A novel strategy for allogeneic stem cell transplantation: perfusion
method plus intra-bone marrow injection of stem cells. Exp Hematol
2003;31(12):1142–6.
85. Palacios R, Golunski E, Samaridis J. In vitro generation of hematopoietic stem
cells from an embryonic stem cell line. Proc Natl Acad Sci USA
1995;92(16):7530–4.
86. Nakano T, Kodama H, Honjo T. In vitro development of primitive and definitive
erythrocytes from different precursors. Science 1996;272(5262):722–4.
87. Wang Y, Yates F, Naveiras O, Ernst P, Daley GQ. Embryonic stem cell-derived
hematopoietic stem cells. Proc Natl Acad Sci USA 2005;102(52):19081–6.
88. Pilat S, Carotta S, Schiedlmeier B, et al. HOXB4 enforces equivalent fates of EScell-derived and adult hematopoietic cells. Proc Natl Acad Sci USA
2005;102(34):12101–6.
89. Eckardt S, Leu NA, Bradley HL, Kato H, Bunting KD, McLaughlin KJ.
Hematopoietic reconstitution with androgenetic and gynogenetic stem cells.
Genes Dev 2007;21(4):409–19.
90. Abramovich C, Humphries RK. Hox regulation of normal and leukemic
hematopoietic stem cells. Curr Opin Hematol 2005;12(3):210–6.
91. Antonchuk J, Sauvageau G, Humphries RK. HOXB4-induced expansion of adult
hematopoietic stem cells ex vivo. Cell 2002;109(1):39–45.
92. Sauvageau G, Thorsteinsdottir U, Eaves CJ, et al. Overexpression of HOXB4 in
hematopoietic cells causes the selective expansion of more primitive
populations in vitro and in vivo. Genes Dev 1995;9(14):1753–65.
93. Buske C, Feuring-Buske M, Abramovich C, et al. Deregulated expression of
HOXB4 enhances the primitive growth activity of human hematopoietic cells.
Blood 2002;100(3):862–8.
94. Davidson AJ, Zon LI. The caudal-related homeobox genes cdx1a and cdx4 act
redundantly to regulate hox gene expression and the formation of putative
hematopoietic stem cells during zebrafish embryogenesis. Dev Biol
2006;292(2):506–18.
95. Wang Y, Yabuuchi A, McKinney-Freeman S, et al. Cdx gene deficiency
compromises embryonic hematopoiesis in the mouse. Proc Natl Acad Sci USA
2008;105(22):7756–61.
96. McKinney-Freeman SL, Lengerke C, Jang IH, et al. Modulation of murine
embryonic stem cell-derived CD41+c-kit+ hematopoietic progenitors by
ectopic expression of Cdx genes. Blood 2008;111(10):4944–53.
97. Lengerke C, McKinney-Freeman S, Naveiras O, et al. The cdx-hox pathway in
hematopoietic stem cell formation from embryonic stem cells. Ann NY Acad Sci
2007;1106:197–208.
98. Riedt T, Ebinger M, Salih HR, et al. Aberrant expression of the homeobox gene
CDX2
in
pediatric
acute
lymphoblastic
leukemia.
Blood
2009;113(17):4049–51.
99. Thoene S, Rawat VP, Heilmeier B, et al. The homeobox gene CDX2 is aberrantly
expressed and associated with an inferior prognosis in patients with acute
lymphoblastic leukemia. Leukemia 2009;23(4):649–55.
100. Rawat VP, Thoene S, Naidu VM, et al. Overexpression of CDX2 perturbs HOX
gene expression in murine progenitors depending on its N-terminal domain
and is closely correlated with deregulated HOX gene expression in human
acute myeloid leukemia. Blood 2008;111(1):309–19.
101. Scholl C, Bansal D, Dohner K, et al. The homeobox gene CDX2 is aberrantly
expressed in most cases of acute myeloid leukemia and promotes
leukemogenesis. J Clin Invest 2007;117(4):1037–48.
102. Mikkola HK, Fujiwara Y, Schlaeger TM, Traver D, Orkin SH. Expression of CD41
marks the initiation of definitive hematopoiesis in the mouse embryo. Blood
2003;101(2):508–16.
103. McKinney-Freeman SL, Naveiras O, Yates F, et al. Surface antigen phenotypes
of hematopoietic stem cells from embryos and murine embryonic stem cells.
Blood.
104. Vodyanik MA, Bork JA, Thomson JA, Slukvin II. Human embryonic stem cellderived CD34+ cells: efficient production in the coculture with OP9 stromal
cells
and
analysis
of
lymphohematopoietic
potential.
Blood
2005;105(2):617–26.
105. Wang L, Menendez P, Shojaei F, et al. Generation of hematopoietic
repopulating cells from human embryonic stem cells independent of ectopic
HOXB4 expression. J Exp Med 2005;201(10):1603–14.
106. Krosl J, Austin P, Beslu N, Kroon E, Humphries RK, Sauvageau G. In vitro
expansion of hematopoietic stem cells by recombinant TAT-HOXB4 protein.
Nat Med 2003;9(11):1428–32.
107. Carotta S, Pilat S, Mairhofer A, et al. Directed differentiation and mass
cultivation of pure erythroid progenitors from mouse embryonic stem cells.
Blood 2004;104(6):1873–80.
108. Fujimoto TT, Kohata S, Suzuki H, Miyazaki H, Fujimura K. Production of
functional platelets by differentiated embryonic stem (ES) cells in vitro. Blood
2003;102(12):4044–51.
109. Eto K, Murphy R, Kerrigan SW, et al. Megakaryocytes derived from embryonic
stem cells implicate CalDAG-GEFI in integrin signaling. Proc Natl Acad Sci USA
2002;99(20):12819–24.
110. Lieber JG, Keller GM, Worthen GS. The in vitro differentiation of mouse
embryonic stem cells into neutrophils. Methods Enzymol 2003;365:129–42.
111. Lieber JG, Webb S, Suratt BT, et al. The in vitro production and characterization
of neutrophils from embryonic stem cells. Blood 2004;103(3):852–9.
112. Tsai M, Tam SY, Wedemeyer J, Galli SJ. Mast cells derived from embryonic
stem cells: a model system for studying the effects of genetic manipulations
on mast cell development, phenotype, and function in vitro and in vivo. Int J
Hematol 2002;75(4):345–9.
113. Hamaguchi-Tsuru E, Nobumoto A, Hirose N, et al. Development and functional
analysis of eosinophils from murine embryonic stem cells. Br J Haematol
2004;124(6):819–27.
114. Cho SK, Zuniga-Pflucker JC. Development of lymphoid lineages from
embryonic stem cells in vitro. Methods Enzymol 2003;365:158–69.
115. de Pooter RF, Cho SK, Zuniga-Pflucker JC. In vitro generation of lymphocytes
from embryonic stem cells. Methods Mol Biol 2005;290:135–47.
116. de Pooter RF, Cho SK, Carlyle JR, Zuniga-Pflucker JC. In vitro generation of T
lymphocytes from embryonic stem cell-derived prehematopoietic
progenitors. Blood 2003;102(5):1649–53.
117. Moore KJ, Fabunmi RP, Andersson LP, Freeman MW. In vitro-differentiated
embryonic stem cell macrophages: a model system for studying
atherosclerosis-associated macrophage functions. Arterioscler Thromb Vasc
Biol 1998;18(10):1647–54.
118. Senju S, Hirata S, Matsuyoshi H, et al. Generation and genetic modification of
dendritic cells derived from mouse embryonic stem cells. Blood
2003;101(9):3501–8.
119. Fairchild PJ, Brook FA, Gardner RL, et al. Directed differentiation of dendritic
cells from mouse embryonic stem cells. Curr Biol 2000;10(23):1515–8.
120. Fairchild PJ, Nolan KF, Waldmann H. Probing dendritic cell function by guiding
the differentiation of embryonic stem cells. Methods Enzymol
2003;365:169–86.
121. Lian RH, Maeda M, Lohwasser S, et al. Orderly and nonstochastic acquisition of
CD94/NKG2 receptors by developing NK cells derived from embryonic stem
cells in vitro. J Immunol 2002;168(10):4980–7.
122. Olsen AL, Stachura DL, Weiss MJ. Designer blood: creating hematopoietic
lineages from embryonic stem cells. Blood 2006;107(4):1265–75.
123. Weiss MJ, Keller G, Orkin SH. Novel insights into erythroid development
revealed through in vitro differentiation of GATA-1 embryonic stem cells.
Genes Dev 1994;8(10):1184–97.
124. Olivier EN, Qiu C, Velho M, Hirsch RE, Bouhassira EE. Large-scale production of
embryonic red blood cells from human embryonic stem cells. Exp Hematol
2006;34(12):1635–42.
125. Qiu C, Olivier EN, Velho M, Bouhassira EE. Globin switches in yolk sac-like
primitive and fetal-like definitive red blood cells produced from human
embryonic stem cells. Blood 2008;111(4):2400–8.
126. Lu SJ, Feng Q, Park JS, et al. Biologic properties and enucleation of red blood
cells from human embryonic stem cells. Blood 2008;112(12):4475–84.
127. Ma F, Ebihara Y, Umeda K, et al. Generation of functional erythrocytes from
human embryonic stem cell-derived definitive hematopoiesis. Proc Natl Acad
Sci USA 2008;105(35):13087–92.
128. Bouhassira EE. Toward the manufacture of red blood cells? Blood
2008;112(12):4362–3.
129. Kaufman DS, Hanson ET, Lewis RL, Auerbach R, Thomson JA. Hematopoietic
colony-forming cells derived from human embryonic stem cells. Proc Natl
Acad Sci USA 2001;98(19):10716–21.
130. Cerdan C, Rouleau A, Bhatia M. VEGF-A165 augments erythropoietic
development from human embryonic stem cells. Blood 2004;103(7):2504–12.
131. Takayama N, Nishikii H, Usui J, et al. Generation of functional platelets from
human embryonic stem cells in vitro via ES-sacs, VEGF-promoted structures
that concentrate hematopoietic progenitors. Blood 2008;111(11):5298–306.
132. Yokoyama Y, Suzuki T, Sakata-Yanagimoto M, et al. Derivation of functional
mature neutrophils from human embryonic stem cells. Blood.
133. Karlsson KR, Cowley S, Martinez FO, Shaw M, Minger SL, James W.
Homogeneous monocytes and macrophages from human embryonic stem
cells following coculture-free differentiation in M-CSF and IL-3. Exp Hematol
2008;36(9):1167–75.
134. Gaur M, Kamata T, Wang S, Moran B, Shattil SJ, Leavitt AD. Megakaryocytes
derived from human embryonic stem cells: a genetically tractable system to
study megakaryocytopoiesis and integrin function. J Thromb Haemost
2006;4(2):436–42.
135. Junt T, Schulze H, Chen Z, et al. Dynamic visualization of thrombopoiesis
within bone marrow. Science 2007;317(5845):1767–70.
136. Woll PS, Grzywacz B, Tian X, et al. Human embryonic stem cells differentiate
into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood.
C. Lengerke, G.Q. Daley / Blood Reviews 24 (2010) 27–37
137. Martin CH, Woll PS, Ni Z, Zuniga-Pflucker JC, Kaufman DS. Differences in
lymphocyte developmental potential between human embryonic stem cell
and umbilical cord blood-derived hematopoietic progenitor cells. Blood
2008;112(7):2730–7.
138. Galic Z, Kitchen SG, Kacena A, et al. T lineage differentiation from
human
embryonic
stem
cells.
Proc
Natl
Acad
Sci
USA
2006;103(31):11742–7.
139. Galic Z, Kitchen SG, Subramanian A, et al. Generation of T lineage cells from
human embryonic stem cells in a feeder free system. Stem cells
2009;27(1):100–7.
37
140. Timmermans F, Velghe I, Vanwalleghem L, et al. Generation of T cells from
human embryonic stem cell-derived hematopoietic zones. J Immunol
2009;182(11):6879–88.
141. Tavian M, Robin C, Coulombel L, Peault B. The human embryo, but not its yolk sac,
generates lympho-myeloid stem cells: mapping multipotent hematopoietic cell fate
in intraembryonic mesoderm. Immunity 2001;15(3):487–95.
142. Lengerke C, Grauer M, Niebuhr NI, et al. Hematopoietic development from
human induced pluripotent stem cells. Ann N Y Acad Sci 2009;1176:219–27.
143. Lengerke C, Daley GQ. Disease models from pluripotent stem cells. Ann N Y
Acad Sci 2009;1176:191–6.