You are on page 1of 18

EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS

1. Gene Regulation, Stem Cells and RUNX1c Regulates Hematopoietic Differentia-


Development Group, Department of
Genomic Oncology, GENYO: Centre for
tion of Human Pluripotent Stem Cells Possibly
Genomics and Oncological Research in Cooperation with Pro-inflammatory Signal-
Pfizer-University of Granada-Junta de
Andaluca, PTS Granada, Granada ing
18016, Spain; 2. Departament of Bio-
chemistry and Molecular Biology I.
Faculty of Science. University of Gra- OSCAR NAVARRO-MONTERO1, VERONICA AYLLON1^, MAR LAMOLDA1,2^,
nada, Granada, Spain; 3. Josep Carreras LOURDES LPEZ-ONIEVA1, ROSA MONTES1, CLARA BUENO3,
Leukemia Research Institute and Bio- ELIZABETH NG4, XIOMARA GUERRERO-CARRENO1, TAMARA
medicine Department, School of Medi-
cine, University of Barcelona, Barcelo- ROMERO1, DAMI ROMERO-MOYA3, ED STANLEY5, ANDREW
na, Spain; 4. Blood Cell Development & ELEFANTY4, VERNICA RAMOS-MEJIA1, PABLO MENENDEZ3,6,7*.
Disease Laboratory. Murdoch Childrens PEDRO J. REAL1,2*
Research Institute. The Royal Chil-
drens Hospital. Parkville, Australia; 5.
Stem Cell Technology Laboratory. Key words. RUNX1c human ESC human PSC hematopoiesis hemato-
Murdoch Childrens Research Institute. endothelial precursors.
The Royal Childrens Hospital. Parkville,
Australia; 6. Instituci Catalana de
Reserca i EstudisAvanats (ICREA),
Barcelona, Spain; 7. Centro de Investi-
ABSTRACT
gacin Biomdica en Red de Cncer
(CIBERONC), ISCIII, Barcelona, Spain. Runx1 is a master hematopoietic transcription factor essential for hemato-
* Correspondence should be ad- poietic stem cell (HSC) emergence, Runx1-deficient mice die during early
dressed to: Pablo Menendez Ph.D, embryogenesis due to the inability to establish definitive hematopoiesis.
MBA, Josep Carreras Leukaemia Re- Here we have used hPSCs as model to study the role of RUNX1 in human
search Institute, School of Medi- embryonic hematopoiesis. Although the three RUNX1 isoforms a, b and c
cine. Barcelona University, Carrer Cas- were induced in CD45+ hematopoietic cells, RUNX1c was the only isoform
anova 143. Barcelona 08036. Spain, induced in hemato-endothelial progenitors (HEPs)/hemogenic endotheli-
Tel. 00 34 935 572 809, E-mail.
um. Constitutive expression of RUNX1c in hESCs enhanced the appearance
pmenendez@carrerasresearch.org,
Pedro J. Real Ph.D., GENyO: Centre for of HEPs, including hemogenic (CD43+) HEPs and promoted subsequent dif-
Genomics and Oncological Research ferentiation into blood cells. Conversely, specific deletion of RUNX1c dra-
Pfizer-Universidad de Granada-Junta matically reduced the generation of hematopoietic cells from HEPs, indicat-
de Andalucia . Parque Tecnolgico de ing that RUNX1c is a master regulator of human hematopoietic develop-
Ciencias de la Salud (PTS) Granada, ment. Gene expression profiling of HEPs revealed a RUNX1c-induced pro-
Avda. de la Ilustracin 114, Granada inflammatory molecular signature, supporting previous studies demon-
18016, Spain, Phone: 00 34 958 715 strating pro-inflammatory signaling as a regulator of HSC emergence. Col-
500 , Email: pedro.real@genyo.es; lectively, RUNX1c orchestrates hematopoietic specification of hPSCs, pos-
^ Equal contribution.
sibly in cooperation with pro-inflammatory signaling. STEM CELLS 2017;
Received September 16, 2016; accept-
ed for publication August 02, 2017; 00:000000
available online without subscription
through the open access option.
AlphaMed Press SIGNIFICANCE STATEMENT
1066-5099/2017/$30.00/0
This article has been accepted for pub-
We decided to elucidate the role of RUNX1 isoforms throughout hematopoi-
lication and undergone full peer review
but has not been through the copyedit- etic specification of human embryonic stem cells (hESCs). We show that
ing, typesetting, pagination and proof- RUNX1c isoform is the first induced in hemato-endothelial progenitors
reading process which may lead to (HEPs), while the other main RUNX1 isoforms are up-regulated latter on in
differences between this version and hESC-derived CD45+ hematopoietic cells. Constitutive expression of RUNX1c
the Version of Record. Please cite this in hESCs improves the generation of HEPs and CD45+ hematopoietic cells,
article as doi: 10.1002/stem.2700 meanwhile specific deletion of this isoform exclusively affected hematopoi-
etic differentiation from HEPs. Furthermore, we show evidence that

STEM CELLS 2017;00:00-00 www.StemCells.com AlphaMed Press 2017


2 RUNX1c promotes human embryonic hematopoiesis

RUNX1c-overexpression in HEPs We believe that our findings advance our knowledge of how human embry-
activates a pro-inflammatory signa- onic hematopoiesis occurs and they could help to improve current protocols
ture in accordance with recent in for hematopoietic differentiation from human pluripotent stem cells.
vivo results.

INTRODUCTION with unlimited cell growth, self-renewal capacity and


pluripotency potential[19, 20]. Directed differentiation
Runt-related transcription factor 1 (RUNX1) also of hPSCs towards hematopoiesis provides a unique cel-
known as AML1 (Acute myeloid leukemia 1) is a mem- lular system for studying the molecular pathways regu-
ber of the Core-binding Factor (CBF) family with a pivot- lating human early hematopoiesis.
al role in hematopoiesis. Runx1-deficient mice die dur- In hESCs, RUNX1c expression remarkably parallels
ing embryonic development between days E11.5 and the expression of hematopoietic markers during differ-
E12.5 due to massive hemorrhages in the central nerv- entiation and is restricted to a subpopulation of CD34+
ous system and the inability to establish definitive hem- cells[10, 21]. Importantly, the overexpression of RUNX1
atopoiesis[1, 2]. Conditional Runx1 knockout mice re- in combination with other transcription factors facili-
vealed its requirement for endothelial-to-hematopoietic tates the conversion of both hPSC-derived HE and ma-
transition (EHT) allowing the formation of intra-arterial ture endothelial cells into HSPCs with engraftment po-
clusters and the generation of HSCs from hemogenic tential[22, 23], indicating a master role of RUNX1 in the
endothelium (HE) but not thereafter[3]. Furthermore, regulation of early human hematopoiesis. Here we have
studies using live-imaging to monitor in vivo the emer- used hPSCs as a cellular model to study the kinetics of
gence of hematopoietic cells from HE have revealed expression of RUNX1 isoforms in human ear-
that Runx1 is necessary for the EHT[46]. Several ly/embryonic hematopoiesis. We show that RUNX1c is
RUNX1 transcripts can be transcribed from two differ- the earliest isoform up-regulated throughout hemato-
ent promoters P1 or distal promoter, and P2 or prox- poietic specification of hPSCs. Importantly, RUNX1c is
imal promoter by alternative splicing[7, 8]. The the only isoform enriched in hemato-endothelial pro-
isoforms RUNX1a, b and c are the most frequently ex- genitors (HEPs), while all three RUNX1 isoforms are ex-
pressed in hematopoietic tissues[710]. RUNX1a and pressed in CD45+ hematopoietic cells. Both gain- and
RUNX1b expression is driven from the P2 promoter loss-of-function experiments demonstrate that RUNX1c
while RUNX1c is transcribed from the P1 promoter[7, regulates the emergence of both hemogenic HEPs and
8]. RUNX1b and c contain a transactivation domain and hematopoietic cells. Finally, transcriptomic analysis of
act as transcription factors, whereas RUNX1a lacks this HEPs identified a pro-inflammatory signature linked to
region and has long been considered a natural domi- RUNX1c-overexpression. Our study provides new in-
nant negative[1113]. sights into the expression pattern of RUNX1 isoforms
There are several discrepancies about the temporal during early human hematopoietic development and
expression and the relevance of each RUNX1 isoform the contribution of RUNX1c isoform in regulating hema-
during embryonic and adult hematopoiesis. In mice, topoietic specification.
Runx1a and Runx1b are dominant in primitive erythro-
cytes at E7.5[14]. Subsequently, both P1 and P2 Runx1 MATERIALS AND METHODS
isoforms are concurrently expressed in hematopoietic
stem and progenitors cells (HSPCs) from yolk sac and hESC culture
Aorta-Gonad-Mesonephros (AGM), whereas P1-driven The hESCs lines AND1, H9, HS181 and HES3 and the iPSC
Runx1 isoforms become predominant in fetal liver (FL) cell line PBMC1-iPS4F1 were maintained undifferentiat-
and throughout adulthood[10, 1416]. However, in ed in a feeder-free culture as previously described[24
zebrafish enhanced green fluorescence protein (EGFP) 26]. HES3 RUNX1c knock-out cells (RUNX1c-/-) were
expression driven by runx1 P1 occurs in erythromyeloid generated by conventional homologous
progenitors and precedes runx1 P2 activation in HSCs recombination[27, 28]. Approval to work with hPSCs
from AGM[17]. Lie-a-ling et al employed mouse ESCs to was obtained from the Spanish National Stem Cell
show that Runx1b regulates the formation of cell clus- Steering Committee.
ters in the HE with hematopoietic potential[18].
Despite considerable advances in animal models, Cloning strategy, lentiviral vectors and trans-
the knowledge about the specific effect of the different
RUNX1 isoforms in human hematopoietic development
duction
is still scarce. Studying human embryonic development Human RUNX1c cDNA (Addgene #12426) was direction-
is challenging due to logistic difficulties for accessing ally subcloned into the intermediate vector KJ-EGFP-2A
embryonic and fetal human tissues. Human pluripotent (kindly provided by Dr. Garca-Perez; GENyO, Granada,
stem cells (hPSCs), including human embryonic stem Spain). Then, the full cassette EGFP-2A-Flag-RUNX1c
cells (hESCs) and induced pluripotent stem cells (iPSCs), was removed and cloned into the PmeI site in the pRRL-
are the only non-transformed human cellular model EF1-PGK-NEO vector (kindly provided by Prof. Trono,

www.StemCells.com AlphaMed Press 2017


3 RUNX1c promotes human embryonic hematopoiesis

Lausanne, Switzerland). HEK-293T cells were tency was analyzed as described[30]. Briefly, 1-2105
transfected with pRRL-EF1-PGK-NEO (Empty Vector: hPSCs were subcutaneously implanted into the flank of
EV) or pRRL-EF1-EGFP-2A-Flag-RUNX1c-PGK-NEO 6-8-week-old NOD/SCID IL2R-/- mice (NSG) (The Jackson
(RUNX1c) together with packaging plasmids (psPAX and Laboratory, Bar Harbor, MA). Teratoma growth was
pMD2.G, from Addgene) by standard calcium- monitored weekly. Mice were sacrificed at 8-10 weeks
phosphate transfection[25]. Supernatants were post implantation and teratomas were removed, fixed
collected 48 hours after transfection and concentrated in formaldehyde, embedded in paraffin, sectioned and
by ultracentrifugation. AND1 and HS181 were infected stained with hematoxylin/eosin or by immunocyto-
overnight on the day of passage with concentrated virus chemistry[31].
supplemented with polybrene (8 g/mL; Sigma-Aldrich).
The viral supernatants were removed on the next day OP9 hematopoietic differentiation system
and infected hESCs were washed and maintained in hESC-OP9 co-cultures were performed as described[25,
culture. Transduced cells were selected with G418 (150 32, 33]. Briefly, OP9 stromal cells were plated in gelatin-
g/mL; Invitrogen) from day 3 to day 15. RUNX1c coated 10-cm dishes in MEM basal medium supple-
expression was confirmed by quantitative real-time mented with 20% non-heat-inactivated FBS for eight
polymerase chain reaction (qRT-PCR) and western days. hESCs grown in Matrigel-coated flasks were pre-
blotting. pared as a suspension of small aggregates using colla-
genase IV followed by gentle scraping in differentiation
Characterization of pluripotency markers in medium (DM: MEM basal medium, 10% non-heat-
hPSCs by flow cytometry inactivated FBS, 100 M monothioglycerol and 50
hPSC colonies were dissociated with TrypLE Express g/mL ascorbic acid). One fifth of the cell suspension
(Invitrogen) and the cell suspension was stained with was plated on top of the OP9 stroma in 10 mL of DM
PE-conjugated TRA-1-60 and SSEA3, APC-conjugated and cells were fed with fresh DM the following day.
TRA-1-81 and SSEA4 and FITC-conjugated OCT-4 anti- From day 4 to 10 of co-culture, a half-volume medium
bodies (BD Bioscience) for 30 minutes. Cells were change was performed every other day. Hematopoietic
washed and stained with 7-aminoactinomycin D (7AAD) specification was analyzed by flow cytometry (at days 6,
(BD Bioscience) and live cells identified by 7AAD exclu- 8 and 10 of co-culture) and CFU assays (at day 8 of co-
sion were analyzed using a FACS Canto II flow cytome- culture). OP9 cells were stained with anti-mouse CD29-
ter[25]. FITC (AbDSerotec, Dsseldorf, Germany) to exclude
mouse cells. The percentage of human HEPs
RNA isolation, RT-PCR and qRT-PCR analysis (CD31+CD34+CD45-), primitive blood cells (CD34+CD45+)
Total RNA was isolated using Trizol (Invitrogen)[25]. and total blood cells (CD45+) was analyzed as
cDNA was generated using the SuperScript First-Strand described[25].
Synthesis System for RT-PCR (Invitrogen) and analyzed
by qRT-PCR using Brilliant III Ultra-Fast SYBR Green Feeder-free hematopoietic differentiation
QPCR Master Mix (Agilent Technologies, La Jolla, CA) on system
the Mx3005P QPCR System (Agilent Technologies). Feeder-free hematopoietic differentiation was per-
Gene expression levels were calculated using the (2-CT formed as previously described[26]. Confluent hPSC
method) and GAPDH was used to normalize data [29]. cultures were disaggregated into single cells with Try-
Where indicated, the relative gene expression was also PLE and plated (0.5-3105 hPSCs) onto six-well plates
calculated using the CT method and GAPDH as refer- coated with 0.5 g/cm2 ColIV in E8 media supplemented
ence gene. Primer sequences are listed in Table S1. with 10M Y27632. Next day, the medium was changed
to IF9S medium containing 50ng/mL BMP4, 50ng/mL
Western blotting FGF2, 15 ng/mL Activin A and 2 mM LiCl. On day 2, me-
hPSCs were dissociated with TrypLE Express and cells dia was changed to IF9S containing 50 ng/mL FGF2 and
lysed in RIPA buffer (Sigma-Aldrich) containing com- 50 ng/mL VEGF, and on day 4 to the media was changed
plete protease inhibitors cocktail (Roche, Basel, Switzer- to Differentiation medium 3 (IF9S containing 50 ng/mL
land) and phosphatase inhibitors (Sigma-Aldrich). Cell FGF2, 50 ng/mL VEGF, 50 ng/mL SCF, 50 ng/mL TPO, 50
lysates were electrophoresed on 12% SDS-PAGE and ng/mL IL6 and 10 ng/mL IL3 [all cytokines from Pepro-
transferred to PVDF membranes. RUNX1c protein was tech]). Cell cultures were maintained in hypoxia (5% O2,
detected with the Odyssey Infrared Imaging System (Li- 5% CO2) for the first 6 days. From day 6 onwards, cells
cor Biosciences, Lincoln, NE) with an anti-RUNX1 anti- were maintained in Differentiation medium 3 in
body (Abcam, Cambridge, MA). An anti--actin antibody normoxia. The frequency of hemogenic (CD43+) and
(Sigma-Aldrich) was used as loading control. non-hemogenic (CD43-) HEPs (CD34+CD31+KDR+CD45-)
were analyzed at day 6 and 8 of development.
In vivo teratoma formation
Animal protocols were approved by the Animal Care
Committee of the University of Granada. In vivo pluripo-

www.StemCells.com AlphaMed Press 2017


4 RUNX1c promotes human embryonic hematopoiesis

Colony-Forming Unit (CFU) assay Gene Expression Hybridization kit and Agilent Whole
Human clonogenic progenitor assays were performed Human Genome Oligo Microarray, 860K (Agilent Tech-
by plating 5104 cells from OP9-hESC co-cultures into nologies). Two independent samples per condition and
methylcellulose H4436 (Stem Cell Technologies, Van- cell line were labeled and hybridized. Candidate genes
couver, Canada). Cells were incubated at 37 C in a hu- with a fold change >2 and a P-value <0.01 were consid-
midified atmosphere and colonies were counted be- ered differentially expressed. Functional analysis and
tween day 10 and 12 using standard morphological cri- canonical pathway studies were performed using the
teria[25, 34]. Panther public web tool (www.pantherdb.org) and In-
genuity Pathway Analysis (IPA) (Ingenuity Systems,
Cell cycle analysis of HEPs www.ingenuity.com) as described[37, 38].
OP9 co-cultures were harvested at day 8 of devel-
opment, fixed in 70% ice-cold ethanol, and stored over- Statistical analysis
night at -20 C. The next day, cells were washed and All data are expressed as meanSEM. Statistical com-
incubated with anti-CD31-FITC and anti-CD34-FITC (Mil- parisons were performed with a paired Students t-test
tenyiBiotec) for 15 minutes. After washing, the cells with the exception of the CFU score analysis, where
were resuspended in propidium iodide (PI) buffer con- Walds test was applied. Values were considered statis-
taining 50 g/mL PI and 100 g/mL RNAase in PBS. Cell tically significant at P<0.05.
cycle distribution was analyzed using a FACSCanto-II
cytometer equipped with Modfit software (Verity Soft- RESULTS
ware House, Topsham, ME)[25, 35].
Expression of RUNX1 isoforms during hema-
Apoptosis analysis of HEPs topoietic differentiation of hPSCs
hESCs cultured over OP9 stromal cells were harvested We first differentiated hPSCs towards the hematopoiet-
at day 8, washed with Annexin V-binding buffer, and ic lineage[25, 32, 33] and analyzed the expression pat-
incubated with anti-human CD31-FITC and anti-CD34- tern of RUNX1a, b and c isoforms by qRT-PCR. HEPs
FITC (MiltenyiBiotec) and Annexin V-APC (Becton Dick- emerged at day 6 of OP9-coculture and then differenti-
inson) antibodies for 20 minutes. After washing, the ate into CD45+ cells from day 8 onwards[25, 37].
cells were resuspended in Annexin V-binding buffer RUNX1a and RUNX1a/b isoforms expression did not
with 7AAD and apoptotic cell death was detected in the increase until day 10 of hematopoietic differentiation,
CD31+CD34+CD45- cell population by flow cytometry whereas the RUNX1c isoform was readily induced at
using a FACSCanto II flow cytometer. day 6, paralleling the appearance of HEPs, and its ex-
pression progressively increased throughout differentia-
Mouse transplantation and analysis of en- tion (Fig. 1a). We then analyzed the expression pattern
graftment of the key hematopoietic transcription factors
NSG mice were housed under sterile conditions. Cord SCL/TAL1, GATA1 and PU.1/SPI1 by qRT-PCR (Fig.1a,
Blood (CB)-derived CD34+ HSPCs (3104 cells in 50 L), S1a). SCL, a master factor for HEPs specification[25] and
as well as EV (1-2.5105 cells in 50 L) or RUNX1c hESC a direct activator of RUNX1[39, 40], was the earliest
derivatives (1-2.5105 cells in 50 L) purified from day 8 induced gene. RUNX1c either preceded or paralleled
differentiating OP9-co-cultures were transplanted in- the induction of GATA-1 and PU.1, both RUNX1
trahepatically into newborn NSG mice[25, 36]. Mouse targets[41, 42].
health was monitored throughout the entire experi- To further characterize the expression pattern of the
ment. Mice were killed 6-8 weeks after transplantation RUNX1 isoforms, we FACS-purified HEPs
and bone marrow (BM), spleen, liver and peripheral (CD34+CD31+CD45-) and CD45+ blood cells at day 8 and
blood were collected and analyzed for human chimer- 10 of differentiation, respectively, and non-
ism. Cells were stained with anti-HLA-ABC-PE and anti- hematopoietic cells (CD31-CD34-CD45-) were also iso-
CD45-APC (BD Bioscience) antibodies to analyze human lated as a control (Fig. 1b, S1b). qRT-PCR analysis re-
chimerism by flow cytometry. vealed that all RUNX1 isoforms are highly expressed in
blood cells, while RUNX1c was the only isoform en-
Gene expression profiling and analysis riched in HEPs in comparison with non-hematopoietic
Undifferentiated hPSCs and 6, 8 and 10-day-old purified cells (Fig. 1c, S1c). Importantly, when HEPs were identi-
HEPs from hESC-EV and hESC-RUNX1c cells were sorted fied more exhaustively as CD45-CD43-CD41-
using a FACS Aria sorter (BD Bioscience) and total RNA CD34+CD31+CD73-CD184-)[16, 43] an identical trend
was isolated using Trizol. High quality RNA was con- was observed, confirming our initial sorting strategy
firmed using the Agilent 2100 Bionalyzer (Agilent Tech- (Fig. 1d). Collectively, RUNX1c is the isoform whose ex-
nologies). RNA samples were labeled (Agilent Low Input pression best parallels early human hematopoietic
Quick Amp Labeling kit, Agilent Technologies) with Cy3 specification in vitro, being the most enriched in HEPs.
following the manufacturers instructions. The hybridi-
zation procedure was accomplished using the Agilent

www.StemCells.com AlphaMed Press 2017


5 RUNX1c promotes human embryonic hematopoiesis

RUNX1c enhances hemato-endothelial speci- expressing hematopoietic derivatives than in EV control


fication of hPSCs and hematopoietic com- cells (Fig. 2g). CFU scoring revealed a slight skew to-
wards macrophage commitment (CFU-M) in RUNX1c-
mitment of HEPs expressing hematopoietic progenitors at the expense of
To further assess the contribution of RUNX1c during granulocyte (CFU-G)/granulocyte-macrophage (CFU-
hematopoietic specification of hPSCs, we over- GM) differentiation (Fig. 2g). Supporting the RUNX1c
expressed RUNX1c in AND1 and HS181 hESCs lines hematopoietic-promoting effects, the hematopoietic
which have good and poor intrinsic hematopoietic dif- transcription factors SCL, GATA1 and PU.1 were also
ferentiation potential, respectively[24, 25] and one iPSC significantly upregulated throughout hematopoietic
cell line[31] with either an empty lentivector (EV) or a differentiation (Fig. 2h and S5a). The ectopic lev-
RUNX1c-expressing lentivector (RUNX1c) (Fig. 2a). After els/expression of RUNX1c was maintained throughout
selection with G418 the transgenic hPSCs showed stable differentiation (Fig. S5b). Collectively, our data indicates
expression of RUNX1c at the mRNA and protein level that RUNX1c over-expression increases the generation
(Figs. 2b,c). Six-to-eight weeks after selection, EV- and of CD43+ hemogenic HEPs, resulting in an increased
RUNX1c-hESCs were analyzed for expression of pluripo- hematopoietic (CD45+) differentiation of hPSCs.
tency markers and in vivo differentiation potential.
RUNX1c-overexpressing hESCs expressed the pluripo-
tency-associated transcription factors OCT4, NANOG
RUNX1c expression does not confer in vivo
and SOX-2 (Figs. S2a,b) and the pluripotency-associated engraftment potential to hPSC-derived hema-
antigens Tra-1-60, Tra-1-81, SSEA-3 and SSEA-4 (Fig. topoietic derivatives
S2b). Both EV and RUNX1c-hESC lines formed teratomas The RUNX1c-mediated enhancement of in vitro hema-
with identical efficiency, latency and histological com- topoietic differentiation prompted us to examine
position with differentiation into tissues representing whether enforced expression of RUNX1cwas sufficient
the three germ layers[25] (Fig. S2c). Thus, RUNX1c to confer in vivo engraftment ability to the hPSC-
overexpression is compatible with pluripotency. derived RUNX1c-expressing hematopoietic derivatives.
Irrespective of the hematopoietic potential of the We transplanted 3104 CB-derived CD34+ HSPCs as a
hPSC line, hematopoietic differentiation on OP9 cells positive control, or 1.5-2.5105 hESC-derived EV and
demonstrated that RUNX1c over-expression significant- RUNX1c hematopoietic derivatives intrahepatically into
ly increased (2- to 12-fold) the generation of HEPs newborn NGS mice[25, 36]. After 8 weeks we analyzed
(CD31+CD34+CD45) (Fig 2d, S3a left panels and S3b). the presence of human hematopoietic cells in the
We next analyzed in more detail the composition of transplanted animals. RUNX1c expression by itself was
HEPs, and we found that RUNX1c over-expression sig- not enough to confer in vivo engraftment potential to
nificantly increased the frequency of CD43+ hemogenic hPSC blood derivatives (Fig. S6), in line with the so far
HEPs at day 5, while the CD43- HEPs were not signifi- reported marginal repopulating ability of hPSC-blood
cantly altered all over differentiation (Fig 2e), suggest- derivatives, and with the idea that a defined set of fac-
ing that constitutive expression of RUNX1c accelerated tors seems necessary to endow in vivo repopulating
the emergence of CD43+ hemogenic HEPs at early stag- ability[22, 23, 25, 37, 4447].
es of hematopoietic differentiation. To rule out an ef-
fect of RUNX1c in either the proliferation or apoptosis RUNX1c targeted deletion reduces EHT
of the emerging HEPs we analyzed HEPs for cell cycle To further investigate the specific contribution of
distribution and apoptosis and found no differences in RUNX1c isoform to the hematopoietic specification of
the proportion of cycling cells (330.3% vs 342.4%) or hPSCs, we harnessed a RUNX1c knock-out hESC line
apoptotic cells (0.3% vs 0.9%) between EV and RUNX1c (RUNX1c-/-) previously generated by conventional ho-
HEPs (Fig. S4), suggesting that ectopic RUNX1c likely mologous recombination[28]. Slight decrease was ob-
promotes specification of HEPs rather than proliferation served in HEP specification in RUNX1c-/- hESCs (Figs 3a,
or survival. S7a left panel and S7b); however, the generation of
The RUNX1c-mediated increase in HEPs specification hematopoietic cells in vitro from these HEPs was signifi-
resulted in a 6-10-fold increase of cantly reduced in RUNX1c-/- cells (Fig 3a, middle and
CD34+CD45+hematopoietic progenitors and total right panel and S7a,b). Importantly, RUNX1c ablation
CD45+ hematopoietic cells (Fig 2f, S3a, middle and right significantly impaired the number of CD45+ blood cells
panels, S3b and S3c). As an indirect measure of the per HEP, confirming a role of RUNX1c in regulating EHT
hematopoietic output of HEPs, we calculated the num- (Fig.S7c). The transcription factors SCL, GATA1 and PU.1
ber of total CD45+ blood cells produced in culture by were also robustly down-regulated in RUNX1c-/- hESC-
HEPs. As shown in Fig S3d, by day 10 of differentiation blood derivatives throughout hematopoietic differentia-
RUNX1c-HEPs produced 3-4-fold higher numbers of tion (Fig.S7d). qRT-PCR confirmed the complete ab-
CD45+ cells than EV-HEPs. We next analyzed the clono- sence of RUNX1c expression throughout blood differen-
genic capacity of EV- and RUNX1c-hematopoietic deriv- tiation in RUNX1c-/- cells (Fig 3b), while RUNX1a and
atives at day 8 of OP9 co-culture, and found that clono- RUNX1a/b isoforms were similarly induced in both
genic potential was 3-8-fold higher in RUNX1c- RUNX1c-/- and RUNX1c+/+ cell lines (Fig. 3c,d). When we

www.StemCells.com AlphaMed Press 2017


6 RUNX1c promotes human embryonic hematopoiesis

assessed the clonogenic potential of the hematopoietic ferentiation, expansion and function of macrophages,
progenitors in CFU assays we found that RUNX1c-/- and granulocytes and their progenitors[50]. Therefore, the
RUNX1c+/+ hESC-hematopoietic derivatives produced activation of the CSF1R signaling pathway could be re-
similar total number of colonies; however, RUNX1c-/- sponsible for the CFU-M/CFU-G skew observed in the
hESC derivatives had an increase in E, a decrease in M clonogenic assays upon RUNX1c over-expression (Fig.
and disappearance of G/GM colonies (Fig 3e), sugges- 2g).
tive of a more primitive hematopoiesis in the absence Pro-inflammatory signals including TNF, IFN and
of RUNX1c. Moreover, SCL, GATA1 and PU.1 were ro- IFN have been recently proposed as key in vivo regula-
bustly down-regulated in RUNX1c-/- hESC-derived CD45+ tors of definitive hematopoiesis in zebrafish and mouse
cells throughout hematopoietic differentiation (Fig. AGM[5154]. We used IPA to visualize the transcrip-
S7c). Taken together, RUNX1c is necessary for the gen- tional network formed by the key inflammatory regula-
eration of CD45+ cells from HEPs. tors and their known targets in RUNX1c HEPs over the
differentiation period. At day 6 of differentiation the
RUNX1c-HEPs show an activated pro- inflammatory regulators are predicted to be inactive
inflammatory transcriptional signature (Fig. S9), but at day 8 the network of inflammatory
To explore the potential mechanisms by which RUNX1c regulators has expanded and it is now predominantly
regulates EHT, we performed gene expression profiling activated, with a complex network including many
using microarrays in HEPs from EV- and RUNX1c-hESCs genes co-regulated by different factors. IFN appears as
at different time points of hematopoietic differentia- the most important factor, based on quantity of target
tion. We confirmed the expression levels of 20 selected genes and co-regulations (Fig. S10). At day 10 the in-
genes by qRT-PCR and found a significant concordance flammatory regulators remain activated; the relative
correlation coefficient (0.76) with the microarray data, importance of IFN now balanced by a high level of TNF
validating the global gene expression data (Fig. S8). To target genes and co-regulations (Fig. S11). Our results
explore the biological meaning of the changes in gene suggest that RUNX1c over-expression induces a gene
expression imposed by RUNX1c overexpression we used signature that recapitulates an activation of different
first the web tool Panther (www.pantherdb.org) and inflammatory signals that converge and form a complex
applied a statistical enrichment test to find the most network at the differentiation time when HEPs are un-
significant gene ontology biological processes altered in dergoing EHT.
RUNX1c-HEPs vs EV-HEPs. Both at day 8 and day 10 of To validate these predictions we performed qRT-PCR
hematopoietic differentiation, the most relevant biolog- analysis of key genes of these inflammatory pathways in
ical processes, ranked by P-value, were associated with purified EV- and RUNX1c-HEPs at days 6, 8 and 10 of
Immune System Process and Immune/Defense Re- hematopoietic differentiation. Among the members of
sponses (Fig. 4a). To confirm this prediction, we used the interferon pathway, we found IFNB1, IFNA2, and
the software Ingenuity Pathway Analysis (IPA) and Interferon Regulatory Factors (IRFs) IRF7 and IRF1 up-
found that the category Hematological System Devel- regulated (Figs. 5a,b and Table S2). Type I interferon
opment & Function was consistently the most signifi- signaling is mediated by Janus Kinase proteins that re-
cant biological process affected in RUNX1c-HEPs and cruit and activate signal transducers and activators of
EV-HEPs (Fig.4b). Using IPA we analyzed in detail the transcription (STATs)[55]. All STAT genes analyzed were
biofunctions altered within this category, and found significantly up-regulated in late (day 10) RUNX1c-HEPs
many functions related to the migration, adhesion and (Fig.5c and Table S2). In addition, RUNX1c over-
differentiation of myeloid and lymphoid cells predicted expression also increased the expression of the NF-B
to be activated (z-score >2) in RUNX1c-over-expressing members NFKB1 and RELA (Fig.5d and Table S2) and
HEPs (Fig. 4c). TLR4 by day 10, while did not up-regulate TNF expres-
Next, we used IPA to identify which upstream regu- sion in HEPs (Fig.5e and Table S2). Interestingly, Blood-
lators could be responsible for the gene expression ChiP analysis reveals that most of these genes are direct
changes associated to RUNX1c over-expression in HEPs. targets of RUNX1 in human HSCs and we found a good
We could distinguish five different gene clusters of up- correlation between the genes modulated by RUNX1c
stream regulators predicted to be activated in RUNX1c over-expression in hESC-derived HEPs and promoter
over-expressing cells (Fig. 4d): i) regulators of macro- occupancy in HSCs (Table S4)[56]. We also sorted out
phage differentiation; ii) megakaryocytic/erythroid reg- HEPs from RUNX1c+/+ and RUNX1c-/- hPSCs and analyzed
ulators; iii) RUNX1 partners and RUNX1-regulated T-cell all pro-inflammatory signaling genes by qRT-PCR. Re-
development; iv) regulators of hematopoietic differen- markably, all were down-regulated or not expressed in
tiation from hPSCs[48, 49]; and v) components of in- RUNX1c-/- purified HEPs (Fig.5f and Table S3). These
flammatory signaling, which represent the biggest clus- data reinforces the hypothesis that RUNX1c over-
ter and agrees with the GO data presented in Figure 4a. expression imposes a gene expression profile that re-
IPA predicted that RUNX1c over-expression activat- flects an activation of pro-inflammatory signaling net-
ed CSF1 and CSF2 from day 6 to 10 and CSF1R at day 8 works which may contribute to in vitro hematopoietic
in HEPs (Fig.5d). CSF1 (M-CSF), CSF2 (GM-CSF) and their differentiation of hPSCs.
receptor CSF1R are necessary for the production, dif-

www.StemCells.com AlphaMed Press 2017


7 RUNX1c promotes human embryonic hematopoiesis

DISCUSSION cytometric analysis within this population confirmed an


increase in CD43+ hemogenic HEPs in RUNX1c over-
The elucidation of the molecular determinants in- expressing cultures. Ectopic over-expression of RUNX1c
ducing hematopoietic specification of hPSCs would facil- further augmented the production of CD45+CD34+
itate the generation of bona fide HSCs for developmen- hematopoietic progenitors and CD45+ blood cells. Cell
tal biology and cell therapy. Our studies demonstrate cycle and apoptosis assays and the outcome of CD45+
that RUNX1c promotes hematopoietic specification, blood cells produced per HEP revealed that RUNX1c is
possibly in cooperation with pro-inflammatory signals. involved in hematopoietic specification rather than pro-
The study of the expression kinetics of the three main liferation and/or survival. We cannot rule out that
RUNX1 isoforms during hPSCs hematopoietic differenti- RUNX1c may be favouring the formation of a
ation revealed that RUNX1c is the first isoform induced subpopulation of early hematopoietic progenitors
upon hematopoietic differentiation, while RUNX1a/b (CD34+CD43+CD45-) within our HEP population. Further
isoforms are up-regulated later on (day 10). In line with dissection of the target population of RUNX1c could
our results, the induction of RUNX1c has been reported elucidate this hypothesis. CFU assays also revealed
to precede RUNX1a/b expression during hematopoietic higher clonogenic potential for RUNX1c-hematopoietic
specification of hESCs using embryoid bodies (EB) dif- derivatives. In addition, RUNX1c over-expression also
ferentiation protocols[10], although Ran et al showed alters the distribution of the types of colonies, slightly
the opposite pattern of RUNX1 induction following the enhancing the CFU-M at the expense of CFU-G and CFU-
spin-EB differentiation system[57]. Likewise, during GM, suggesting a role in monocytic differentiation. In
mouse ESC hematopoietic differentiation, P2- contrast, RUNX1a over-expression in hPSCs provokes an
transcribed Runx1 isoforms emerged earlier that P1- increase in the number of GEMM and erythroid colonies
transcribed Runx1c[14, 15]. Thus, regulation of RUNX1 at the expense of the rest[57], that together with the
isoforms appears subject to species-specific differences, enhanced number of hematopoietic progenitors
and peculiarities associated with hematopoietic differ- suggest a role of RUNX1a in proliferation of the HSPCs
entiation methods and hPSC lines used. In addition, our compartment rather than differentiation. Interestingly,
qPCR results also show that SCL is up-regulated before RUNX1c over-expression induced the expression of the
RUNX1c, GATA1 emerges concomitantly to RUNX1c; key hematopoietic transcription factors SCL, GATA1 and
while PU. 1 is induced later on after blood cells appear- PU.1. The regulation of GATA1[41] and PU.1[42] by
ance. RUNX1 has been previously reported, however the
In our differentiation system RUNX1c is the only iso- especific activation by RUNX1c was unknown so far.
form enriched in HEPs and the highest up-regulated More surprising is the induction of SCL by RUNX1c, a
isoform in hESC-derived CD45+ cells in comparison with known upstream regulator of RUNX1, in contrast to
non-hematopoietic derivatives, confirming previous previous publications[17, 41]. One posibility will be that
results from our and other labs[10, 58]. In genetically- RUNX1c up-regulates a different isoform of SCL not
modified mESCs containing reporter genes controlled implicated in establishing the HE, as suggested in
by P1 and P2 promoters, Sroczynska et al showed that zebrafish[60]. These results could be reproduced in two
the HE expresses RUNX1b while definitive hematopoiet- hESCs lines with high and low hematopoietic potential
ic cells mainly expressed RUNX1c[15]. Very recently, and one iPSC cell line. Despite overexpressing different
Elefantys laboratory, using spin-EB differentiation pro- ectopic levels of RUNX1c, all lines displayed very similar
tocols showed that induction of RUNX1c occurs after RUNX1c-mediated hematopoietic phenotype, further
emergence of HE [28, 43]. These discrepancies can likely validating a role for RUNX1c regardless the cellular
be attributed to species-specific differences, different background and hematopoietic output.
differentiation strategies and the read-out assays for Similar to many studies using hPSC-derived hemato-
measuring RUNX1c up-regulation[10, 15, 57]. For ex- poietic cells from non-manipulated or genetically-
ample, in contrast to our OP9 co-culture, EB-based modified hPSCs [25, 37, 4447], RUNX1c-hematopoietic
blood differentiation protocols require the addition of derivatives also failed to engraft into newborn NSG
hematopoietic cytokines such as IL3, which might by- mice. Similarly, murine ESC-derived hematopoietic pro-
pass RUNX1-mediated transcription regulators[59]. Fur- genitors transduced with RUNX1c failed to confer en-
thermore, early-acting hematopoietic cytokines might graftment in transplantation assays[10]. By contrast,
modify the finely tuned regulation of P1 and P2 pro- Ran et al showed short-term myeloid-restricted multi-
moters, leading to abnormal expression of RUNX1 tran- lineage engraftment in 33% of mice transplanted with
scripts. RUNX1a-expressing hESC-derived CD34+CD45+ cells[57].
Given its expression pattern during hematopoietic However, RUNX1 was very recently found to be a mas-
differentiation of hPSCs, we hypothezyed that RUNX1c ter factor that in combination with other transcription
would contribute to HEPs specification and further factors facilitates the conversion of both hPSC-derived
blood (CD45+) generation. Using a gain-of function HE and mature endothelial cells into HSPCs with en-
approach, we found that RUNX1c over-expression was a graftment potential[22, 23], further emphasizing the
booster of HEP emergence and blood production, master role of RUNX1 in human hematopoietic for-
something not described so far. A more detailed flow mation. Of note, RUNX1a over-expression might alter

www.StemCells.com AlphaMed Press 2017


8 RUNX1c promotes human embryonic hematopoiesis

normal hematopoietic development because its expres- predicted the activation of several set of genes with
sion is significantly up-regulated in acute leukemia pa- different functionalities. The first cluster included genes
tients and its constitutive expression in murine BM cells controlling macrophage functions, specifically CSF1,
contributes to leukemogenesis[13]. Available evidence CSF2 and CSF1R. This activation could explain the slight-
suggests that RUNX1a and RUNX1c isoforms play differ- ly higher number of CFU-M in RUNX1c hematopoietic
ent roles throughout mammalian hematopoietic devel- progeny, a previously unrecognized function of RUNX1c
opment. in monocyte/macrophage development.
To corroborate the relevance of RUNX1c during hu- RUNX1c-mediated hematopoietic specification of
man embryonic hematopoiesis we next completed OP9 hESCs involved a pro-inflammatory transcriptional sig-
differentiation with RUNX1c-/- hESCs generated by con- nature, consistent with recent studies in zebrafish and
ventional homologous recombination[28]. Intriguingly, mice demonstrating that pro-inflammatory signaling is a
specific deletion of RUNX1c did not impact HEP specifi- positive regulator of hematopoietic development[51
cation, but profoundly impaired the commitment of 54]. Our in silico analysis of the gene expression profile
HEPs into CD45+ hematopoiesis. In contrast, Elefantys imposed by RUNX1c showed that this profile is con-
laboratory has recently shown that RUNXc deletion sistent with an activation of several pro-inflammatory
does not affect the emergence of HEPs or CD45+ blood regulators, but we have no evidence that RUNX1c di-
cells using a spin-EB differentiation protocol[28]. The rectly regulates their expression. Expression analysis of
unaffected HEP compartment could be explained if: 1) RUNX1c-expressing and RUNX1c-/- HEPs confirmed that
RUNX1c is not required for its formation or 2) there is a several members of the type I interferon signaling
deficiency in a specific subpopulation that we are not pathway and the NF-B members NFKB1 and RELA were
able to detect because the limited resolution of the modulated by RUNX1c. Interestingly, BloodChiP analysis
panel of antibodies used. Intriguingly, RUNX1c-/-- reveals that most of these genes are direct targets of
hematopoietic derivatives displayed similar CFU poten- RUNX1 in human HSCs[56]. However, Dr Menendezs
tial to RUNX1c+/+ counterparts but distinct colony type lab has demonstrated that the addition of individual
distribution (absence of G/GM, reduced M and in- pro-inflammatory cytokines throughout hematopoietic
creased E) was observed in RUNX1c-/- hESCs compared differentiation in both EBs and OP9 systems is not suffi-
to RUNX1+/+ counterparts. Interestingly, Ng et al did not cient to potentiate definitive hematopoietic specifica-
observe differences in the CFU potential between tion of hPSCs in vitro[61].These results indicate that in
RUNX1c-/- and RUNX1c+/- hESCs[28]. Our results suggest vitro hPSC hematopoietic differentiation does not reca-
that RUNX1c deletion could block the progression from pitulate the microenvironmental signals present in vivo
primitive, where only erythrocytes, macrophages and in the embryo. For instance, primitive neutrophils origi-
megakaryocytes cells can be generated, to definitive nated in the yolk sac are necessary for the establish-
hematopoiesis in OP9-based hematopoietic differentia- ment of definitive HSCs in vivo in the AGM[51], but the-
tion systems. In line with this hypothesis Runx1c -/- mice se cells are absent in in vitro experiments. Further work
had altered definitive hematopoietic progenitor cell is required to biochemically and functionally confirm
number and only the primitive E, M and megakaryo- that RUNX1c orchestrates hematopoietic specification
cyte-CFU were obtained[15, 16]. Alternatively, it cannot of hPSCs in cooperation with pro-inflammatory signal-
be ruled out that cytokine cocktails present in spin-EB ing.
differentiation systems and methylcellulose cultures
might bypass RUNX1c deletion in vitro by activating CONCLUSION
signaling pathways downstream of RUNX1c. In OP9 co-
culture system the absence of those cytokines enhances We show that RUNX1c is an important transcription
the relevance of RUNX1c in the transition from HEPs to factor regulating human early blood specification, pos-
CD45+ blood cells. In addition, RUNX1c deficiency sibly in cooperation with pro-inflammatory signals. The
caused a drastic reduction in SCL, GATA1 and PU.1 ex- resolution of the spatio-temporal expression of these
pression, confirming RUNX1c over-expression results, immune regulators and the integration with other
and placing RUNX1c as a putative transcription regula- known hematopoietic regulators could serve as a plat-
tor of these 3 hematopoietic transcription factors. Col- form to efficiently activate a bona fide hematopoietic
lectively, our results suggest that RUNX1c is dispensable transcriptional program in HSCs derived from hPSCs.
for the generation of HEPs but is necessary for the gen-
eration of CD45+ blood cells from hPSCs[16, 21, 43]. ACKNOWLEDGMENTS
Finally, to decipher the molecular mechanisms trig-
gered by RUNX1c that enhance hematopoietic differen-
We thank the Andalusian Bioinformatics Platform (PAB)
tiation of hESCs, we performed gene expression profil-
at the University of Malaga (www.scbi.uma.es) for
ing analysis of EV- and RUNX1c-overexpressing HEPs at
providing access to Ingenuity Pathway Analysis soft-
different time points. As expected, RUNX1c activates a
ware, Dr. J.L. Garca-Perez (GENyO, Granada, Spain) for
molecular signature related with the Hematological
the 2A-peptide vector (KJ-2A vector) and technical sup-
System Development & Function. A deeper analysis
port, Dr. D. Trono (Ecole Polytechnique Federale de
allowed us to discover that RUNX1c over-expression

www.StemCells.com AlphaMed Press 2017


9 RUNX1c promotes human embryonic hematopoiesis

Lausanne, Lausanne, Switzerland) for the pRRL-EF1- by the MINECO (JCI_2012_12666). LL-O was a Marie
PGK-NEO vector, Dr. M. Alarcon-Riquelme and Dr. C. Curie Intra-European Fellow (FP7-MC-IEF-623806).
Maranon (GENyO, Granada, Spain) and Dr. L Espinosa
(IMIM, Barcelona, Spain) for providing reagents and Dr. AUTHOR CONTRIBUTIONS
M. Martinez-Bueno (GENyO, Granada, Spain) for his
assistance with statistical analysis. This work was fund- O.N.-M. performed most experiments, analyzed the
ed by the Junta de Andaluca/FEDER (SAS-111244 and data and wrote the manuscript. R.M., M.M. L.L-O., X.G.-
P10-CTS-6406 to P.J.R.), FIS/FEDER (PI10/00449) to P.M. C., T.R. and D.R.-M. contributed to the experiments and
(CP09/0063, PI12/01596 and CPII15/00018) to P.J.R. data analysis. V.A. and V.R-M contributed the experi-
(CP07/00059, PI11/00119 and PI14/01191) to C.B. and mental design, data analysis and wrote the manuscript.
(CP12/03175 and PI14/01412) to V.R-M., the MINECO C.B. contributed the experimental design and data anal-
(PLE-2009-0111 and SAF2013/43065) to P.M. and RYC- ysis. E.N., E.S. and A.E. provided important materials.
2015-18382 to P.J.R., Spanish Association Against Can- P.M. and P.J.R. conceived and supervised the project
cer Foundation (CI110023 to P.M. and CI15 to C.B.) and and wrote the manuscript.
Health Canada H9080-144541 to P.M. P.M also
acknowledges the support from the European Research DISCLOSURE OF CONFLICTS OF INTEREST
Council (CoG-2014-646903) and the Obra Social La
Caixa-Fundacin Josep Carreras. RM is supported by the The authors declare no potential conflicts of interest.
Postdoctoral Subprogramme Juan de la Cierva founded

REFERENCES functional effects in adult hematopoietic from human blastocysts. SCIENCE


stem cells. EXP. HEMATOL. 2010;38(5):403 1998;282(5391):11457.
16. 20 Takahashi K, Tanabe K, Ohnuki M, et al.
1 Okuda T, Deursen J van, Hiebert SW, et al.
11 Tanaka T, Tanaka K, Ogawa S, et al. An Induction of pluripotent stem cells from adult
AML1, the target of multiple chromosomal
acute myeloid leukemia gene, AML1, human fibroblasts by defined factors. CELL
translocations in human leukemia, is essential
regulates hemopoietic myeloid cell 2007;131(5):86172.
for normal fetal liver hematopoiesis. CELL
differentiation and transcriptional activation 21 Ferrell PI, Xi J, Ma C, et al. The RUNX1
1996;84(2):32130.
antagonistically by two alternative spliced +24 Enhancer and P1 Promoter Identify a
2 Wang Q, Stacy T, Miller JD, et al. The
forms. EMBO J. 1995;14(2):34150. Unique Subpopulation of Hematopoietic
CBFbeta subunit is essential for CBFalpha2
12 Okuda T, Takeda K, Fujita Y, et al. Progenitor Cells Derived from Human
(AML1) function in vivo. CELL
Biological characteristics of the leukemia- Pluripotent Stem Cells. STEM CELLS
1996;87(4):697708.
associated transcriptional factor AML1 2015;33(4):11301141.
3 Chen MJ, Yokomizo T, Zeigler BM, et al.
disclosed by hematopoietic rescue of AML1- 22 Sugimura R, Jha DK, Han A, et al.
Runx1 is required for the endothelial to
deficient embryonic stem cells by using a Haematopoietic stem and progenitor cells
haematopoietic cell transition but not
knock-in strategy. MOL. CELL. BIOL. from human pluripotent stem cells. NATURE
thereafter. NATURE 2009;457(7231):88791.
2000;20(1):31928. 2017;545(7655):432438.
4 Bertrand JY, Chi NC, Santoso B, et al.
13 Liu X, Zhang Q, Zhang D-E, et al. 23 Lis R, Karrasch CC, Poulos MG, et al.
Haematopoietic stem cells derive directly
Overexpression of an isoform of AML1 in Conversion of adult endothelium to
from aortic endothelium during development.
acute leukemia and its potential role in immunocompetent haematopoietic stem
NATURE 2010;464(7285):10811.
leukemogenesis. LEUKEMIA 2009;23(4):739 cells. NATURE 2017;545(7655):439445.
5 Kissa K, Herbomel P. Blood stem cells
45. 24 Ramos-Mejia V, Melen GJ, Sanchez L, et
emerge from aortic endothelium by a novel
14 Bee T, Liddiard K, Swiers G, et al. al. Nodal/Activin signaling predicts human
type of cell transition. NATURE
Alternative Runx1 promoter usage in mouse pluripotent stem cell lines prone to
2010;464(7285):1125.
developmental hematopoiesis. BLOOD CELLS. differentiate toward the hematopoietic
6 Boisset J-C, Cappellen W van, Andrieu-
MOL. DIS. 43(1):3542. lineage. MOL. THER. 2010;18(12):217381.
Soler C, et al. In vivo imaging of
15 Sroczynska P, Lancrin C, Kouskoff V, et 25 Real PJ, Ligero G, Ayllon V, et al.
haematopoietic cells emerging from the
al. The differential activities of Runx1 SCL/TAL1 regulates hematopoietic
mouse aortic endothelium. NATURE
promoters define milestones during specification from human embryonic stem
2010;464(7285):11620.
embryonic hematopoiesis. BLOOD cells. MOL. THER. 2012;20(7):144353.
7 Miyoshi H, Ohira M, Shimizu K, et al.
2009;114(26):527989. 26 Uenishi G, Theisen D, Lee J-H, et al.
Alternative splicing and genomic structure of
16 Bee T, Swiers G, Muroi S, et al. Tenascin C promotes hematoendothelial
the AML1 gene involved in acute myeloid
Nonredundant roles for Runx1 alternative development and T lymphoid commitment
leukemia. NUCLEIC ACIDS RES.
promoters reflect their activity at discrete from human pluripotent stem cells in
1995;23(14):27629.
stages of developmental hematopoiesis. chemically defined conditions. STEM CELL
8 Levanon D, Glusman G, Bangsow T, et al.
BLOOD 2010;115(15):304250. REPORTS 2014;3(6):107384.
Architecture and anatomy of the genomic
17 Lam EYN, Chau JYM, Kalev-Zylinska ML, 27 Davis RP, Grandela C, Sourris K, et al.
locus encoding the human leukemia-
et al. Zebrafish runx1 promoter-EGFP Generation of human embryonic stem cell
associated transcription factor RUNX1/AML1.
transgenics mark discrete sites of definitive reporter knock-in lines by homologous
GENE 2001;262(12):2333.
blood progenitors. BLOOD recombination. CURR. PROTOC. STEM CELL
9 Miyoshi H, Shimizu K, Kozu T, et al. t(8;21)
2009;113(6):12411249. BIOL. 2009;Chapter 5:Unit 5B.1 1.1-34.
breakpoints on chromosome 21 in acute
18 Lie-a-ling M, Marinopoulou E, Li Y, et al. 28 Ng ES, Azzola L, Bruveris FF, et al.
myeloid leukemia are clustered within a
RUNX1 positively regulates a cell adhesion Differentiation of human embryonic stem
limited region of a single gene, AML1. PROC.
and migration program in murine hemogenic cells to HOXA+ hemogenic vasculature that
NATL. ACAD. SCI. U. S. A.
endothelium prior to blood emergence. resembles the aorta-gonad-mesonephros.
1991;88(23):104314.
BLOOD 2014;124(11):e11-20. NAT. BIOTECHNOL. 2016;34(11):11681179.
10 Challen GA, Goodell MA. Runx1 isoforms
19 Thomson JA, Itskovitz-Eldor J, Shapiro 29 Livak KJ, Schmittgen TD. Analysis of
show differential expression patterns during
SS, et al. Embryonic stem cell lines derived Relative Gene Expression Data Using Real-
hematopoietic development but have similar

www.StemCells.com AlphaMed Press 2017


10 RUNX1c promotes human embryonic hematopoiesis

Time Quantitative PCR and the 2CT 38 Toscano MG, Navarro-Montero O, stimulating factors (Gcsfs): 2 paralogous
Method. METHODS 2001;25(4):402408. Ayllon V, et al. SCL/TAL1-mediated cytokines and their roles in hematopoietic
30 Gutierrez-Aranda I, Ramos-Mejia V, transcriptional network enhances development and maintenance. BLOOD
Bueno C, et al. Human induced pluripotent megakaryocytic specification of human 2013;122(24):391828.
stem cells develop teratoma more efficiently embryonic stem cells. MOL. THER. 51 Espn-Palazn R, Stachura DL, Campbell
and faster than human embryonic stem cells 2015;23(1):15870. CA, et al. Proinflammatory signaling regulates
regardless the site of injection. STEM CELLS 39 Nottingham WT, Jarratt A, Burgess M, et hematopoietic stem cell emergence. CELL
2010;28(9):156870. al. Runx1-mediated hematopoietic stem-cell 2014;159(5):107085.
31 Montes R, Romero T, Cabrera S, et al. emergence is controlled by a Gata/Ets/SCL- 52 Li Y, Esain V, Teng L, et al. Inflammatory
Generation and characterization of the regulated enhancer. BLOOD signaling regulates embryonic hematopoietic
human iPSC line PBMC1-iPS4F1 from adult 2007;110(13):41884197. stem and progenitor cell production. GENES
peripheral blood mononuclear cells. STEM 40 Landry J-R, Kinston S, Knezevic K, et al. DEV. 2014;28(23):2597612.
CELL RES. 2015;15(3):614617. Runx genes are direct targets of Scl/Tal1 in 53 Sawamiphak S, Kontarakis Z, Stainier
32 Vodyanik MA, Bork JA, Thomson JA, et the yolk sac and fetal liver. BLOOD DYR. Interferon gamma signaling positively
al. Human embryonic stem cell-derived 2008;111(6):300514. regulates hematopoietic stem cell
CD34+ cells: efficient production in the 41 Yokomizo T, Hasegawa K, Ishitobi H, et emergence. DEV. CELL 2014;31(5):64053.
coculture with OP9 stromal cells and analysis al. Runx1 is involved in primitive 54 He Q, Zhang C, Wang L, et al.
of lymphohematopoietic potential. BLOOD erythropoiesis in the mouse. BLOOD Inflammatory signaling regulates
2005;105(2):617626. 2008;111(8):40754080. hematopoietic stem and progenitor cell
33 Choi KD, Vodyanik M, Slukvin II. 42 Huang G, Zhang P, Hirai H, et al. PU.1 is a emergence in vertebrates. BLOOD
Hematopoietic differentiation and production major downstream target of AML1 (RUNX1) 2015;125(7):1098106.
of mature myeloid cells from human in adult mouse hematopoiesis. NAT. GENET. 55 Capobianchi MR, Uleri E, Caglioti C, et al.
pluripotent stem cells. NAT PROTOC 2008;40(1):5160. Type I IFN family members: similarity,
2011;6(3):296313. 43 Ditadi A, Sturgeon CM, Tober J, et al. differences and interaction. CYTOKINE
34 Montes R, Ayllon V, Gutierrez-Aranda I, Human definitive haemogenic endothelium GROWTH FACTOR REV. 2015;26(2):10311.
et al. Enforced expression of MLL-AF4 fusion and arterial vascular endothelium represent 56 Chacon D, Beck D, Perera D, et al.
in cord blood CD34+ cells enhances the distinct lineages. NAT. CELL BIOL. BloodChIP: a database of comparative
hematopoietic repopulating cell function and 2015;17(5):58091. genome-wide transcription factor binding
clonogenic potential but is not sufficient to 44 Kaufman DS. Toward clinical therapies profiles in human blood cells. NUCLEIC ACIDS
initiate leukemia. BLOOD using hematopoietic cells derived from RES. 2014;42(Database issue):D172-7.
2011;117(18):47464758. human pluripotent stem cells. BLOOD 57 Ran D, Shia W-J, Lo M, et al. RUNX1a
35 Bueno C, Montes R, Melen GJ, Ramos- 2009;114(17):35133523. enhances hematopoietic lineage commitment
Mejia V, Real PJ, Aylln V, Sanchez L, Ligero G, 45 Slukvin II. Hematopoietic specification from human embryonic stem cells and
Gutierrez-Aranda I, Fernndez AF, Fraga MF, from human pluripotent stem cells: current inducible pluripotent stem cells. BLOOD
Moreno-Gimeno I, Burks D, Plaza-Calonge advances and challenges toward de novo 2013;121(15):288290.
Mdel C, Rodrguez-Manzaneque JC MP, generation of hematopoietic stem cells. 58 Real PJ, Navarro-Montero O, Ramos-
Bueno C, Montes R, et al. A human ESC model BLOOD 2013;122(25):40354046. Mej??a V, et al. The role of RUNX1 isoforms in
for MLL-AF4 leukemic fusion gene reveals an 46 Ramos-Meja V, Navarro-Montero O, hematopoietic commitment of human
impaired early hematopoietic-endothelial Aylln V, et al. HOXA9 promotes pluripotent stem cells. BLOOD
specification. CELL RES 2012;22(6):9861002. hematopoietic commitment of human 2013;121(26):52502.
36 Navarro-Montero O, Romero-Moya D, embryonic stem cells. BLOOD 59 Uchida H, Zhang J, Nimer SD. AML1A
Montes R, Ramos-Meja V, Bueno C, Real PJ 2014;124(20):306575. and AML1B can transactivate the human IL-3
MP, Navarro-Montero O, Romero-Moya D, et 47 Vo LT, Daley GQ. De novo generation of promoter. J. IMMUNOL. 1997;158(5):2251
al. Intrahepatic transplantation of cord blood HSCs from somatic and pluripotent stem cell 8.
CD34+ cells into newborn NOD/SCID- sources. BLOOD 2015;125(17):26418. 60 Zhen F, Lan Y, Yan B, et al. Hemogenic
IL2R(null) mice allows efficient multi-organ 48 Yu QC, Hirst CE, Costa M, et al. APELIN endothelium specification and hematopoietic
and multi-lineage hematopoietic engraftment promotes hematopoiesis from human stem cell maintenance employ distinct Scl
without accessory cells. CLIN. IMMUNOL. embryonic stem cells. BLOOD isoforms. DEVELOPMENT
2012;145(2):8991. 2012;119(26):624354. 2013;140(19):397785.
37 Aylln V, Bueno C, Ramos-Meja V, et al. 49 Smith BW, Rozelle SS, Leung A, et al. The 61 Giorgetti A, Castao J, Bueno C, et al.
The Notch ligand DLL4 specifically marks aryl hydrocarbon receptor directs Proinflammatory signals are insufficient to
human hematoendothelial progenitors and hematopoietic progenitor cell expansion and drive definitive hematopoietic specification of
regulates their hematopoietic fate. LEUKEMIA differentiation. BLOOD 2013;122(3):37685. human HSCs in vitro. EXP. HEMATOL.
2015;29(8):174153. 50 Stachura DL, Svoboda O, Campbell CA, 2017;45:8593.e2.
et al. The zebrafish granulocyte colony-

See www.StemCells.com for supporting information available online. STEM


CELLS ; 00:000000

www.StemCells.com AlphaMed Press 2017


11 RUNX1c promotes human embryonic hematopoiesis

FIGURE 1. Expression of RUNX1 isoforms in HEPs and blood cells derived from hESCs.
A) qRT-PCR analysis showing expression levels of endogenous RUNX1 isoforms (RUNX1a, RUNX1a/b and RUNX1c),
SCL, GATA1 and PU.1 throughout hematopoietic differentiation. B) Schematic representation of FACS sorting of HEPs
and hematopoietic cells (left panel), and representative flow cytometry dot plots showing how HEPs
(CD31+CD34+CD45-), hematopoietic progenitors (CD34+CD45+) and total blood cells (CD45+) are identified (right pan-
el). C) qRT-PCR showing expression levels of endogenous RUNX1a, RUNX1a/b and RUNX1c isoforms in isolated cell
populations in one representative hPSC line. (D) qRT-PCR analysis of endogenous RUNX1 isoforms in highly-purified
FACS-sorted HEPs (CD45-CD43-CD41-CD34+CD31+CD73-CD184-) in one representative hPSC line. Data represents
meanSEM of 3 independent experiments. Statistical significance was assessed with t-Students test. *P<0.05,
**P<0.01 and ***P<0.001.

www.StemCells.com AlphaMed Press 2017


12 RUNX1c promotes human embryonic hematopoiesis

www.StemCells.com AlphaMed Press 2017


13 RUNX1c promotes human embryonic hematopoiesis

FIGURE 2. Enforced expression of RUNX1c augments hematopoietic specification from hPSCs. A) Schematic repre-
sentation of the lentiviral vectors used to overexpress RUNX1c and the EV control. LTR, long terminal repeat; EF1,
elongation factor 1; PGK, phosphoglycerate kinase; NEO, Neomycin resistance cassette; EGFP, enhanced green fluo-
rescent protein; 2A, 2A self-cleaving peptide; Flag, Flag epitope. B) Representative qRT-PCR analysis of RUNX1c. Ex-
pression levels are shown normalized to GAPDH, used as reference gene. Data represents meanSEM of 3 independ-
ent experiments. C) Representative western blot detection of RUNX1c protein in hPSCs. -actin is used as a loading
control.
D) Kinetics of HEPs specification in two different hPSCs lines (HS181 and AND1) transduced with the empty vector
(EV) or RUNX1c-expressing vector (RUNX1c). E) Kinetics at day 5 and 8 of hematopoietic differentiation for CD43+
(hemogenic) and CD43- (non-hemogenic) HEPs in empty vector (EV) or RUNX1c-overexpressing (RUNX1c) hPSCs. F)
Emergence of hematopoietic progenitors (CD34+CD45+) and total blood cells (CD45+) from two different hESCs lines.
G) Number and colony distribution of CFUs per 10000 human cells and colony type distribution. H) Expression levels
of endogenous SCL, GATA1 and PU.1 in hPSC-derived cells. Data represents meanSEM of 3 independent experi-
ments. Statistical significance was assessed with t-Students test except for the CFU scoring data where the Walds
test was used. n.s. (non-significant),*P<0.05, **P<0.01 and ***P<0.001.

www.StemCells.com AlphaMed Press 2017


14 RUNX1c promotes human embryonic hematopoiesis

www.StemCells.com AlphaMed Press 2017


15 RUNX1c promotes human embryonic hematopoiesis

FIGURE 3. RUNX1c specific deletion drastically diminishes EHT.


A) Kinetics of hematopoietic specification from RUNX1c+/+ and RUNX1c -/- hPSCs. B) Expression levels of endogenous
RUNX1c in RUNX1c-/- hematopoietic cells confirming RUNX1c specific deletion in RUNX1c-/- hESCs. ND, non-detected.
C-D) qRT-PCR analysis showing similar induction of RUNX1a (C) and RUNX1a/b (D) endogenous expression in
RUNX1c+/+ and RUNX1c-/- hPSCs. E) Number of CFUs per 10000 human cells and colony type distribution. Data repre-
sents meanSEM of 3-6 independent experiments. Statistical significance was assessed with t-Students test except
for the CFU scoring data where the Walds test was used. *P<0.05, **P<0.01 and ***P<0.001.

www.StemCells.com AlphaMed Press 2017


16 RUNX1c promotes human embryonic hematopoiesis

FIGURE 4. Enforced expression of RUNX1c in HEPs induces a pro-inflammatory signaling signature. A) Top gene on-
tology biological processes (obtained by Panther) enriched in genes differentially expressed in RUNX1c-HEPs vs EV-
HEPs at day 8 and 10 of development. B) Top 15 biological functions of genes differentially expressed in RUNX1c-HEPs
as compared with EV-HEPs at day 6, 8 and 10 using IPA. C) Predicted biofunctions within the Hematological System
Development & Function activated in RUNX1-HEPs at day 6, 8 and 10. D) Highlight of the upstream regulators pre-
dicted to be activated (orange) or repressed (blue) by RUNX1-expressing HEPs at day 6, 8 and 10 of OP9-hESC cocul-
ture.

www.StemCells.com AlphaMed Press 2017


17 RUNX1c promotes human embryonic hematopoiesis

FIGURE 5. Enforced expression of RUNX1c in HEPs regulates the expression of pro-inflammatory mediators. A-E)
qRT-PCR analysis of several genes in RUNX1c-overexpressing HEPs at day 6, 8 and 10. Relative expression is shown
normalized to EV-HEPs. GAPDH is used as an internal control. qRT-PCR analysis of a set of IFNs(A), IRFs (B), STATs (C),
NFKB1 and RELA (D) and TLR4 and TNF (E) predicted to be activated by RUNX1cover-expression in HEPs at day 6, 8
and 10. F) qRT-PCR analysis of a set of IFNs, IRFs, STATs, NFKB1, RELA, TNF and TLR4 in RUNX1c-/- sorted HEPs at day 6
of hematopoietic differentiation. Relative expression is shown normalized to RUNX1c+/+ sorted HEPs. GAPDH is used
as an internal control. Data represents meanSEM of 3 independent experiments.

www.StemCells.com AlphaMed Press 2017


18 RUNX1c promotes human embryonic hematopoiesis

Graphical Abstract

The transcription factor RUNX1c regulates human early blood specification. RUNX1c, possibly in cooperation with
pro-inflammatory signals, enhances the emergence blood derivatives from hPSC cultures.

www.StemCells.com AlphaMed Press 2017

You might also like