ArticlePDF Available

Identification of Multipotent Progenitors that Emerge Prior to Hematopoietic Stem Cells in Embryonic Development

Authors:

Abstract and Figures

Hematopoiesis in the embryo proceeds in a series of waves, with primitive erythroid-biased waves succeeded by definitive waves, within which the properties of hematopoietic stem cells (multilineage potential, self-renewal, and engraftability) gradually arise. Whereas self-renewal and engraftability have previously been examined in the embryo, multipotency has not been thoroughly addressed, especially at the single-cell level or within well-defined populations. To identify when and where clonal multilineage potential arises during embryogenesis, we developed a single-cell multipotency assay. We find that, during the initiation of definitive hematopoiesis in the embryo, a defined population of multipotent, engraftable progenitors emerges that is much more abundant within the yolk sac (YS) than the aorta-gonad-mesonephros (AGM) or fetal liver. These experiments indicate that multipotent cells appear in concert within both the YS and AGM and strongly implicate YS-derived progenitors as contributors to definitive hematopoiesis.
Content may be subject to copyright.
Stem Cell Reports
Ar ticle
Identification of Multipotent Progenitors that Emerge Prior to Hematopoietic
Stem Cells in Embryonic Development
Matthew A. Inlay,
1,5,
*
Thomas Serwold,
2
Adriane Mosley,
1
John W. Fathman,
1,3
Ivan K. Dimov,
1
Jun Seita,
1
and Irving L. Weissman
1,4
1
Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford University, Stanford, CA 94305, USA
2
Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
3
Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
4
Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
5
Present address : Sue and Bill Gross Stem Cell Research Center, Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine,
CA 92697, USA
*Correspondence: minlay@uci.edu
http://dx.doi.org/10.1016/j.stemcr.2014.02.001
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/3.0/).
SUMMARY
Hematopoiesis in the embryo proceeds in a series of waves, with primitive erythroid-biased waves succeeded by definitive waves, within
which the properties of hematopoietic stem cells (multilineage potential, self-renewal, and engraftability) gradually arise. Whereas self-
renewal and engraftability have previously been examined in the embryo, multipotency has not been thoroughly addressed, especially at
the single-cell level or within well-defined populations. To identify when and where clonal multilineage potential arises during embryo-
genesis, we developed a single-cell multipotency assay. We find that, during the initiation of definitive hematopoiesis in the embryo, a
defined population of multipotent, engraftable progenitors emerges that is much more abundant within the yolk sac (YS) than the aorta-
gonad-mesonephros (AGM) or fetal liver. These experiments indicate that multipotent cells appear in concert within both the YS and
AGM and strongly implicate YS-derived progenitors as contributors to definitive hematopoiesis.
INTRODUCTION
In the mammalian blood system, all mature blood lineages,
including erythrocytes, platelets, and all innate and adap-
tive immune cells, are generated from hematopoietic
stem cells (HSCs). In adults, HSCs reside almost exclusively
in the bone marrow. In the embryo, however, hematopoi-
esis is characterized by distinct yet overlapping waves of
blood development, appearing in multiple sites, with prim-
itive erythroid-biased waves succeeded by definitive waves
with increasing lineage potential and functionality. The
functional properties that define adult HSCs do not appear
at once during development but emerge gradually over the
course of several days.
In the mouse embryo, the first blood-forming cells
appear approximately 7.5 days into gestation (embryonic
day [E] 7.5) within the blood islands that line the extraem-
bryonic yolk sac (YS) (Moore and Metcalf, 1970). These
‘primitive’ blood-forming cells appear to be lineage-
restricted, form primarily large nucleated erythrocytes,
and express embryonic globins (Palis et al., 1999). They
also lack the ability to engraft when transplanted intrave-
nously into lethally irradiated adult mice, a hallmark prop-
erty of fully functional adult bone marrow HSCs (Mu
¨
ller
et al., 1994). After the establishment of a circulatory system
at e8.5, ‘definitive’ erythromyeloid progenitors appear
within the YS (Palis et al., 1999), the placenta (PL)
(Alvarez-Silva et al., 2003), and the embryo proper (EP).
The earliest intraembryonic hematopoietic progenitors
are found within the para-aortic splanchnopleura (p-Sp),
which develops into the aorta-gonad-mesonephros
(AGM) that contains the dorsal aorta (Cumano et al.,
1996; Godin et al., 1993, 1995; Medvinsky et al., 1993).
Hematopoietic progenitors with the ability to self-renew
appear within the YS and AGM at e9.0 and appear within
the fetal liver (FL) a day or two later (Yoder and Hiatt,
1997). e9.5 YS cells lack the ability to home to the bone
marrow when transplanted into adult mice, but their
long-term self-renewal activity can be revealed in vivo by
transplantation into the liver or facial vein of sublethally
irradiated newborn mice (Yoder and Hiatt, 1997; Yoder
et al., 1997a, 1997b) or alternatively by first coculturing
with reaggregated AGM tissue (Taoudi et al., 2008)oron
the OP9 bone marrow stromal line (Rybtsov et al., 2011),
indicating that progenitors residing within the YS can
mature into functional HSC. These embryonic progenitors
were thought to be precursors to HSCs, or ‘pre-HSCs,’ and
whereas not precisely defined, pre-HSCs expressed markers
associated with endothelial (VE-cadherin) and hematopoi-
etic (CD41 then CD45) cells (Rybtsov et al., 2011). At e10.5,
fully functional HSCs have been isolated from the dorsal
aorta of the AGM region (Mu
¨
ller et al., 1994), the extraem-
bryonic YS, PL (Gekas et al., 2005), and from the vitelline
and umbilical vessels (de Bruijn et al., 2000). At e11.5,
HSCs are also found within the FL, which then becomes
the predominant site of hematopoiesis until the formation
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 457
A
B
C
D
EF
Figure 1. Development of an In Vitro Clonal Multipotency Assay
(A) Lentiviral constructs used to generate a tetracycline-inducible Dll1 OP9 stromal line (ODT). Construct C329 (top) drives constitutive
expression of the tetracycline-inducible transactivator rtTA3. Construct C388 allows induction of Dll1 expression when the transactivator
is activated in the presence of doxycycline (DOX).
(B) Strategy for clonal assay. ODT stroma is plated the day prior to cell sorting (day 1). At day 0, cells are clone sorted directly onto ODT
stroma and cytokines are added (SCF, TPO, EPO, Flt3L, IL-7, and IL-15). At day 3, hematopoietic colonies are counted. At day 5, cells are fed
and Flt3L, IL-7, IL-15, and DOX (1 mg/ml) are added. At day 9 or 10, colonies are harvested and analyzed by FACS.
(C) Representative hematopoietic output at day 10 of culture of unsorted e12.5 FL. Representative examples of multipotent output from
single-cell cultures can be found in Figure S1.
(D–F) Testing multipotency assay on adult bone marrow (BM) stem/progenitor cells. (D) Sort gates for KIT
+
Lin
SCA-1
+
(KLS) cells and
subpopulations of KLS, including HSCs (CD34
, SLAMF1
+
) and three fractions of multipotent progenitors (MPP) are shown. (E) The
(legend continued on next page)
458 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
of a bone-marrow cavity several days later ( Gekas et al.,
2005; Mu
¨
ller et al., 1994). Thus, the maturation of blood-
forming cells takes place in discrete steps and likely at
several different sites.
A fundamental unresolved question is whether definitive
hematopoietic cells derive directly from the primitive
precursors that first appear in the YS blood islands (Moore
and Metcalf, 1970) or instead emerge separately from a
hematoendothelial precursor in the dorsal aorta called
hemogenic endothelium (Dzierzak and Medvinsky, 1995;
Nishikawa et al., 1998). A large body of evidence supports
the de novo generation of HSCs within the dorsal aorta,
including ex vivo tissue explants of the dorsal aorta prior
to circulation (Cumano et al., 1996, 2001; Medvinsky and
Dzierzak, 1996). Also, time-lapse imaging of AGM sections
in culture reveals the emergence of hematopoietic clusters
from within the luminal wall of the dorsal aorta in mice,
which express several HSC markers, such as KIT, SCA-1,
and CD41 (Boisset et al., 2010). Definitive hematopoietic
progenitors also exist within the YS (Huang and Auerbach,
1993; Kumaravelu et al., 2002). However, early studies
could not exclude the possibility that such progenitors
originated elsewhere and then migrated to the YS. Evidence
supporting a distinct YS origin of definitive hematopoiesis
comes from lineage-tracing experiments that used a
Runx1 Cre-estrogen receptor (ER) reporter to exclusively
label YS-derived hematopoietic cells; subsequent anal-
ysis of these mice revealed labeling of adult HSCs
(Samokhvalov et al., 2007). Similarly, inducible rescue of
Runx1 expression in Runx1 knockout embryos demon-
strated that definitive hematopoiesis could only be rescued
at the developmental stages when Runx1 expression was
restricted to the YS (Tanaka et al., 2012). In Ncx1
/
embryos, which lack a heartbeat and thus circulation, all
hematopoietic cells are found within the YS and PL
prior to embryonic lethality at e10.5 (Lux et al., 2008;
Rhodes et al., 2008). Additionally, transplantation of YS
cells from e8 to e9 allogeneic donors into the YS cavities
of e8 to e9 hosts in utero led to YS blood-island engraft-
ment and, when analyzed several months after birth,
gave rise to donor-derived spleen colony-forming myeloer-
ythroid cells and thymic and peripheral T cells (Weissman
et al., 1977, 1978). Therefore, maturation of early YS stem/
progenitors to adult HSC was demonstrated, but the
cellular identity of HSC precursors, their sites of matura-
tion, and the molecular mechanisms involved remain a
mystery.
Identification of the key populations that give rise to
each wave of embryonic hematopoiesis may provide
critical insights into the relationship between primitive
and definitive hematopoiesis. However, the surface
markers used to isolate adult HSCs and downstream stages
have proven unreliable for identifying the equivalent
embryonic populations (Cumano and Godin, 2007). As a
result, the cells that initiate each hematopoietic wave
remain poorly defined. Multipotency in hematopoiesis
refers to the ability of a progenitor to give rise to all blood
lineages: myeloid cells including erythrocytes, platelets,
monocytes, tissue macrophages, and granulocytes, as well
as lymphocyte lineages including T, B, natural killer (NK),
and dendritic cells. Multipotency also distinguishes defini-
tive hematopoiesis from more primitive cells with limited
lineage potential. Conclusive evidence of multipotency
requires a single-cell assay to prevent the false positives
that may occur when mixtures of lineage-committed
progenitors collectively produce all lineages. In the em-
bryo, multipotency was first observed within the AA4.1
+
p-Sp population at e9.5 (Godin et al., 1995). YS AA4.1
+
WGA
hi
cells possess myeloid and lymphoid potential
in vitro by e10.5 and robustly by e11.5, though this was
not demonstrated clonally (Huang and Auerbach, 1993).
Thus, whereas much is known about the timing of appear-
ance of progenitor potentials within the early embryo, the
clonal identities of the cells with multipotent potentials are
less clear. Therefore, we sought to define the earliest clon-
ally multipotent cells and to determine their distribution
within the sites of early hematopoiesis.
RESULTS
Development of an In Vitro Clonal Multilineage Assay
In order to identify clonally multipotent populations
throughout embryonic development, we developed a
single-cell multipotency assay (Figure 1). The bone-
marrow-derived OP9 stromal line can be used to generate
most hematopoietic lineages in culture, with the notable
exception of T lymphocytes (Kodama et al., 1994). Alterna-
tively, a modified OP9 stromal line that expresses Dll1
(OP9-DL1) can promote T lineage development but
inhibits B cell development (Schmitt and Zu
´
n
˜
iga-Pflu
¨
cker,
2002). Because of the importance of detecting lymphocyte
potential in distinguishing definitive hematopoietic cells
from their primitive myeloid-restricted counterparts, we
distribution of lineage potential of colonies from adult BM KLS cells, showing cells that produced a single branch (MegE in red, GM in green,
and L in blue), two branches (MegE + GM in orange, GM + L in yellow, and MegE + L in purple), and all three branches (MegE + GM + L in
white). Cells that produced lineages from all three branches (white) were scored as multipotent. Cells that gave rise to colonies that did not
survive to day 10 are shown in black. The number of hematopoietic colonies scored (n) is indicated. (F) Distribution of lineage potential in
colonies derived from sorted KLS subpopulations. Note that only HSCs gave multipotent (white) readout.
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 459
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
sought a method to generate both B cells and T cells from a
single cell within a single well. We previously published an
assay whereby common lymphocyte progenitors could be
cultured and expanded on OP9 stroma without commit-
ting to either the B or T cell lineages (Karsunky et al.,
2008). We therefore generated an OP9 stromal line with
an inducible Dll1 expression cassette (Figure 1A). In this
line, which we call ODT, addition of doxycycline (DOX)
rapidly induces surface expression of Dll1 , driving uncom-
mitted lymphocyte progenitors to the T cell lineage,
whereas B-committed progenitors resist T cell induction
and continue to produce B cells (M.A.I. and I.L.W., unpub-
lished data). Hematopoietic progenitors were plated on
ODT stroma along with a combination of hematopoietic
cytokines (SCF, TPO, EPO, Flt3L, interleukin [IL]-7, and
IL-15); DOX was added 4 to 5 days into the culture (Fig-
ure 1B), leading to eight hematopoietic lineages (erythro-
cytes, platelets, macrophages, granulocytes, dendritic cells,
natural killer cells, B cells, and T cells), representing the
three major branches of hematopoiesis: megakaryocyte/
erythrocyte (MegE), granulocyte/monocyte (GM), and
lymphoid (L) (Figure 1C). Representative fluorescence-
activated cell sorting (FACS) plots of single-cell-derived
colonies scored as multipotent can be found in Figure S1
available online. To validate this assay, we sorted adult
bone marrow (BM) KIT
+
Lin
SCA-1
+
(KLS) cells, a popula-
tion that contains HSCs and multipotent progenitors, and
analyzed their clonal lineage potential (Figures 1D and 1E).
Individual KLS cells produced a variety of different lineage
outcomes: some cells yielded only a single lineage, others
generated multiple lineages, and about 15% of colonies
produced lineages representing each of the three major
hematopoietic branches. These colonies we scored as mul-
tipotent. We next separated the KLS population into four
subsets based on expression of CD34, SLAMF1, and FLK2,
markers that identify HSCs and other multipotent progen-
itor populations, and repeated the assay (Figures 1D and
1F). Whereas each population could collectively produce
all lineages, we found that only the HSC population
(CD34
FLK2
SLAMF1
+
KLS) contained multipotent cells,
despite the fact that all KLS subpopulations are known to
be multipotent in vivo. Thus, this assay can reveal multipo-
tency of individual cells, although not all multipotent cells
are revealed.
Nearly All Hematopoietic Colony-Forming Cells
Reside in the KIT
+
CD43
+
Fraction
In our assay conditions, hematopoietic stem and progeni-
tor cells give rise to distinct colonies by day 3, and thus
we could also use this assay to identify markers for these
cells. We initially screened eight markers (KIT, CD43,
CD34, CD41, SCA-1, CD45, AA4.1, and SLAMF1) from
YS, AGM, and FL tissues from e9.5 to e12.5 to identify
which markers could consistently enrich or deplete for
colony-forming activity (Figure S2). For most markers,
colony-forming activity was found in both the positive
and negative fractions. For example, whereas the hemato-
poietic marker CD41 could enrich hematopoietic colony-
forming ability at e9.5, it became downregulated at later
time points, resulting in colony-forming activity within
the CD41
fraction (Figure S2A). However, two markers,
KIT and CD43, nearly uniformly marked all colony-form-
ing cells in all tissues from e9.5 to e12.5.
Emergence of Multipotent KLS Cells during
Embryogenesis
Based on our initial screen, we restricted the search for
multipotent cells to subpopulations within the KIT
+
CD43
+
fraction. We stained tissues with a variety of anti-
bodies to identify candidate populations within the KIT
+
CD43
+
fraction that we could test for clonal multilineage
potential (Figures 2 and S3). In the adult, all multipotent
stem and progenitors are found in the KLS fraction. We
identified a similar population that is KIT
+
CD43
+
and
SCA-1
+
, which appears in e9.5 YS and AGM and e11.5 FL
(Figure 2). This population is highly enriched for colony-
forming activity (between 35% and 50% of plated cells
gave rise to colonies) and, as a population, gives rise to all
lineages in vitro (data not shown). We refer to this embry-
onic population as KLS (KIT
+
Lin
SCA-1
+
) to reflect its
similarity to the analogous KLS population in adult BM,
although embryonic KLS cells also are defined by expres-
sion of CD43, and the only required lineage marker
for negative gating is the red-blood-cell marker TER119
(Figure S3A).
In adult BM, myeloid progenitors are found within the
KIT
+
SCA-1
(MYP) fraction, which contains common
myeloid progenitors (CMP), granulocyte-monocyte pro-
genitors (GMP), and megakaryocyte-erythrocyte pro-
genitors (MEP) ( Akashi et al., 2000). In the embryo, we
identified a similar MYP population, from which emerged
CMP and then MEP and GMP, both by surface phenotype
and in vitro lineage potential (Figure S3B; data not shown).
CD11A Expression Divides Embryonic KLS into Two
Distinct Fractions
The surface markers SLAMF1, FLK2, and CD34 are known
to subdivide adult KLS into HSCs and downstream multi-
potent progenitors (Figure 1D). We examined these
markers in embryonic KLS and found that their expression
levels varied from time point to time point and from tissue
to tissue, making the use of these markers unreliable to sub-
divide embryonic KLS prior to e11.5 (Figure S4). However,
we did find that the KLS population could be consistently
subdivided based on expression of CD11A (Figure 3A).
CD11A (Itgal) is a component of the leukocyte adhesion
460 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
complex lymphocyte function associated 1 (LFA-1), an
integrin involved in several immune system functions
including leukocyte trafficking and lymphocyte activation
(Hibbs et al., 1991; Hogg et al., 2011). In a related
manuscript, we show that adult BM HSCs can also be
subdivided based on CD11A expression and only the
CD11A
fraction contains functional HSCs (J.W.F., M.A.I.,
Nathaniel B. Fernhoff, J.S., and I.L.W., unpublished data).
CD11A
KLS cells uniformly expressed high levels of the
endothelial adhesion molecule VE-cadherin (VE-CAD),
whereas CD11A
+
KLS cells expressed variable levels of VE-
cadherin (Figure 3A). We also examined the expression of
several other markers including the endothelial-associated
markers TIE2 and endoglin and the hematopoietic markers
CD45 and MAC-1 (Figure S5). Whereas the expression
patterns of these markers were less consistent than
CD11A and VE-cadherin across all tissues and time points,
in general we found that endothelial markers were
more highly expressed at earlier time points and on
CD11A
KLS cells, whereas hematopoietic markers were
more highly expressed at later time points and on
CD11A
+
KLS cells.
We next tested CD11A
and CD11A
+
KLS cells for clonal
multilineage potential (Figures 3B and 3C). In adult BM,
the KLS population contains all multipotent cells (Figures
1D–1F). At the population level, both CD11A
and
CD11A
+
KLS cells were able to give rise to all lineages,
including lymphoid B, T, and NK cells (data not shown).
However, only the CD11A
fraction was able to simulta-
neously give rise to MegE, GM, and lymphoid cells at the
single-cell level, indicating the presence of multipotent
cells within this population (Figures 3B and 3C). We found
multipotent CD11A
KLS cells in all tissues examined,
from e9.5 to e11.5. Conversely, in the CD11A
+
KLS
010
3
10
4
10
5
0
10
3
10
4
10
5
1.99
25
010
3
10
4
10
5
0
10
3
10
4
10
5
12.4
47.6
010
3
10
4
10
5
0
10
3
10
4
10
5
4.9319.7
010
3
10
4
10
5
0
10
3
10
4
10
5
1.43
55.6
010
3
10
4
10
5
0
10
3
10
4
10
5
15.546.5
010
3
10
4
10
5
0
10
3
10
4
10
5
7.38
10.5
010
3
10
4
10
5
0
10
3
10
4
10
5
3.53
39.5
010
3
10
4
10
5
0
10
3
10
4
10
5
24.1
43.5
010
3
10
4
10
5
0
10
3
10
4
10
5
11.5
10.6
010
3
10
4
10
5
0
10
3
10
4
10
5
14.6
5.21
Adult BM
e9.5 e10.5 e11.5
AGM
FL
YS
SCA-1
KIT
KLS MYP
Gate: CD43
+
TER119
-
Figure 2. KIT versus SCA-1 Expression of CD43
+
Cells in YS, AGM, and FL from e9.5 to e11.5
KIT (y axis) and SCA-1 (x axis) expression of CD43
+
adult BM is shown in the upper left corner. Gates of KIT
+
SCA-1
+
‘KLS’ and KIT
+
SCA-1
myeloid progenitors ‘MYP’ are shown, with the percent of KLS and MYP populations among CD43
+
TER119
cells indicated. See Figure S3A
for CD43/TER119 plots and gates.
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 461
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
A
B
C
(legend on next page)
462 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
population, we found bipotent MegE/GM and GM/L col-
onies but never all three branches at once, regardless of
time point or tissue (Figure 3C). Thus, the CD11A
KLS
cells population contains all the multipotent cells from
e9.5 to e11.5 and appears in both extra- and intraem-
bryonic regions around e9.5.
YS Contains the Most Multipotent Cells from E9.5 to
E11.5
Our assay also allowed us to estimate the numbers of multi-
potent progenitors in each tissue from e9.5 to e11.5, based
on colony-forming activity (Figure 4A). We found that,
from e8.5 to e10.5, the YS contained the majority of
Figure 3. Clonal Analysis of CD11A
and CD11A
+
KLS
(A) VE-cadherin (VE-CAD; y axis) versus CD11A (x axis) expression of embryonic KLS from e9.5 to e11.5.
(B) Percent of multipotent colonies from clone-sorted CD11A
and CD11A
+
KLS subsets. The percent of multipotent colonies arising from
adult BM KLS is indicated in black and was duplicated from Figure 1E. For e9.5 and e10.5, CD11A
and CD11A
+
KLS cells derived from the
entire embryo proper (EP) were analyzed. The number of multipotent colonies observed out of total colonies scored is listed in parentheses.
(C) Lineage distribution of CD11A
and CD11A
+
KLS colonies. The distribution of lineages that resulted from clone-sorted CD11A
and
CD11A
+
KLS cells onto ODT stroma from e9.5 to e11.5 is shown. A description of the scoring system can be found in the legend to Figure 1E.
The percentage of colonies scored as multipotent is shown as white bars and is identical to the data shown in (B).
AB
CD
E
Figure 4. Numeric Analysis of Hemato-
poietic Progenitor Cells in Embryonic
Development
(A) Absolute number of colony-forming
cells per embryo from YS (blue squares),
AGM (red downward triangles), and FL
(green diamonds) from e8.5 to e11.5. At
e8.5 and e9.5, the whole embryo proper (EP;
orange upright triangles) was cultured
instead of AGM and FL. Unsorted tissues
were plated onto ODT stroma at serial
dilutions and colonies counted at day 3. The
number of litters analyzed (n) for each time
point is indicated.
(B–D) The absolute numbers of CD11A
KLS
(B), CD11A
+
KLS (C), and MYP (D) cells were
calculated for YS (blue), AGM (red), and FL
(green) from e9.5, e10.5, and e11.5. The
numbers shown are per tissue, per embryo.
Error bars are SD. The asterisk (*) in (B)
indicates a statistically significant differ-
ence (unpaired t test) between the absolute
number of CD11A
KLS cells in the YS and
AGM at e10.5 (p < 0.002). The number of
litters analyzed (n) for each tissue (YS in
blue, AGM in red, FL in green) is indicated.
(E) Estimation of the absolute number of
clonally multipotent cells per tissue per
embryo. These numbers were generated by
multiplying the absolute number of CD11A
KLS (from B) times the percent of multi-
potent cells contained within (from Fig-
ure 3B).
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 463
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
A
BC
(legend on next page)
464 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
progenitors, peaking around 1,000 colony-forming cells at
e10.5 and e11.5. By e11.5, the FL had surpassed the YS
and became the predominant source of hematopoiesis
from then on. At all time points examined, the AGM con-
tained the fewest colony-forming cells.
We also estimated the absolute number of CD11A
KLS,
CD11A
+
KLS, and MYP (Lin
CD43
+
KIT
+
SCA-1
) popula-
tions in each tissue (Figures 4B–D). Collectively, these three
populations contain all KIT
+
CD43
+
cells and, thus, likely
all hematopoietic stem/progenitors. We found that the
YS contained approximately 300 CD11A
KLS cells per
embryo at e10.5, roughly six times as many as in the
AGM at this time point (Figure 4B). Furthermore,
CD11A
KLS numbers appeared to decrease in the YS at
e11.5 and increase in the AGM and FL. At e11.5, the FL
contained the most CD11A
+
KLS and MYP (Figures 4C
and 4D). Because CD11A
KLS cells contain all multipotent
activity at e11.5, this suggests that the bulk of the cells that
initially seed the FL are CD11A
+
KLS and MYP, which we
hypothesize are downstream of CD11A
KLS.
With our estimates of the absolute number of CD11A
KLS cells (Figure 4B) and the frequency of multipotent cells
within each tissue (Figure 3B), we could estimate the
absolute number of multipotent cells (Figure 4E). We
found that e10.5 YS contains the most multipotent cells
of all tissues and time points examined, and at e11.5, the
YS still contained more multipotent cells than AGM or
FL. Taken together, our examination of the absolute num-
ber of CD11A
KLS and multipotent cells suggests that
the YS is a major source of multipotent cells at the stages
when fully functional HSCs first appear in embryonic
development.
EPCR Marks a Subset of CD11A
KLS and Can Partially
Enrich for Multipotency
The protein C receptor EPCR (CD201; Procr) has previously
been reported to be expressed in adult HSCs, as well as
embryonic FL e12.5 HSCs (Balazs et al., 2006; Iwasaki
et al., 2010). We found that EPCR is expressed on a subset
of KLS cells as early as e9.5 and remains expressed
throughout gestation (Figure 5A). EPCR expression corre-
lates with high VE-CAD expression and lower CD11A
expression. Using the clonal multipotency assay, we
observed multipotent colony formation from both EPCR
+
and EPCR
subsets of CD11A
KLS cells, though multipo-
tency appeared greater in the EPCR
+
subfraction overall
(Figures 5B, 5C, and S1). We also identified CD11A
and
CD11A
+
KLS cells in the placenta (Figure S6), a region
known to be a niche for hematopoietic stem cells (Gekas
et al., 2005; Ottersbach and Dzierzak, 2005), and included
placental populations in the analysis. Although we did
not observe multipotency in e10.5 PL CD11A
KLS, we
did find it within the EPCR
+
CD11A
KLS at e11.5 in the
PL. Taken together, our data suggest that EPCR can partially
enrich for multipotent cells, though not all multipotent
cells are EPCR
+
.
Lineage-Tracing KLS Cells In Vivo
To better understand the lineage relationship between
CD11A
and CD11A
+
KLS cells, we examined two line-
age-tracing reporter mouse strains (Figure S7). CD11A
KLS cells generally express higher levels of TIE2 than
CD11a
+
KLS cells (Figure S5A); thus, we crossed Tie2
Cre
mice (Kisanuki et al., 2001) to the mT/mG reporter strain
(Muzumdar et al., 2007), which permanently switches
from Tomato to GFP fluorescence in any cell that expresses
Cre (Figure S7A). In the Tie2
cre
3 mT/mG embryos, we
found that all KLS cells were GFP
+
and thus derived from
TIE2-expressing precursors (Figure S7B). However, other
cell types express TIE2, including hemogenic endothelium,
so we next crossed VE-cadherin
CreER
mice (Monvoisin et al.,
2006; Zovein et al., 2008) to mT/mG reporters (Figures
S7C–S7E). In this cross, when tamoxifen is administered,
all cells that express VE-cadherin during the 24 hr window
when tamoxifen is active will permanently express GFP in
themselves and in their progeny. Because VE-cadherin
expression is associated with earlier time points and more
multipotent cells, we hypothesized that VE-CAD
+
cells
would precede VE-CAD
cells. We injected tamoxifen
into pregnant females and examined embr yos at 24, 48,
and 72 hr after injection to monitor the distribution of
GFP-expressing KLS cells. We found that, at 24 hr, the
majority of labeled KLS cells were VE-CAD
+
(both
CD11A
and CD11A
+
), but at 48 and 72 hr, the fraction
of labeled VE-CAD
KLS cells increased and the frequency
of labeled CD11A
KLS cells decreased, consistent with
the notion that VE-CAD
+
KLS cells are giving rise to
VE-CAD
KLS cells. Whereas neither reporter conclusively
demonstrates that CD11A
KLS cells give rise to CD11A
+
KLS cells, our data support our contention that early
Figure 5. Clonal Multipotent Analysis of EPCR
+
and EPCR
KLS Subsets
(A) VE-CAD (y axis) versus EPCR (x axis) expression of KLS cells in YS, AGM, FL, and PL from e9.5 to e11.5. Gates and percentages of EPCR
+
and EPCR
KLS cells are indicated. Nearly all EPCR
+
cells were CD11A
(data not shown).
(B and C) Percent of multipotent colonies in EPCR
+
and EPCR
KLS subsets in e10.5 (B) and e11.5 (C) CD11A
and CD11A
+
KLS cells. For
CD11A
+
KLS, only VE-CAD
+
cells were analyzed. ND, not detected; NA, not analyzed. The number of multipotent colonies observed out of
total colonies scored is indicated in parentheses. FACS analyses of representative multipotent colonies from e11.5 EPCR
+
CD11A
KLS cells
can be found in Figure S1.
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 465
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
hematopoietic progenitors that express endothelial-
associated markers generally precede those that express
hematopoietic markers.
Multipotency Moves into the CD11A
+
KLS Subfraction
at E12.5
By e12.5, the FL is by far the dominant site of hemato-
poiesis and is known to produce fully functional HSCs by
transplantation (Morrison et al., 1995). When we exam-
ined e12.5 FL KLS, nearly all (95%) of KLS cells were
CD11A
+
(Figure 6A). When we examined both CD11A
and CD11A
+
KLS fractions for clonal multilineage poten-
tial, we now found multipotency in the CD11A
+
KLS
fraction (Figures 6B–6D). Interestingly, only the VE-CAD
lo
subset of CD11A
+
KLS cells showed multipotency, whereas
the VE-CAD
CD11A
+
KLS subset, which represents over
A
C
B
D
Figure 6. Clonal Analysis of E12.5 FL KLS Subsets
(A) Gating of CD11A
and CD11A
+
KLS cells in e12.5 FL. Only CD43
+
CD34
+
TER119
cellsare shown. Percentof cells within each gate are shown.
(B) Distribution of clonal lineage potential in e12.5 FL EPCR
+
and EPCR
CD11A
and CD11A
+
KLS cells. The legend is described in Figure 1E.
Only CD11A
KLS and VE-CAD
+
CD11A
+
KLS cells were analyzed.
(C) Percent of multipotent colonies in e12.5 FL CD11A
and CD11A
+
KLS subsets. The number of multipotent colonies observed out of total
colonies scored is indicated in parentheses.
(D) Estimation of the absolute number of multipotent colonies per embryo in e12.5 FL CD11A
and CD11A
+
KLS cells, calculated as in
Figure 4E.
466 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
3
/
4
of CD11A
+
KLS cells, lacked multipotency. By e12.5, the
multipotent CD11A
KLS population was exceedingly rare
compared to the robustly expanding CD11A
+
KLS
population (Figures 6A and 6D). Thus, it appears that the
primary source of multipotent cells shifts from CD11A
to CD11A
+
KLS cells by e12.5.
Only CD11A
KLS Cells Produce All Hematopoietic
Lineages In Vivo
The in vitro multipotency assay demonstrates that, from
e9.5 to e11.5, only the CD11A
KLS population contains
clonal multilineage potential. We next compared the in vivo
lineage potential of CD11A
and CD11A
+
KLS cells by
competitive transplantation (Figure 7). We sorted CD11A
and CD11A
+
KLS cells from cyan fluorescent protein
(CFP)
+
and CFP
embryos and mixed CFP
+
CD11A
KLS
cells with CFP
CD11A
+
KLS cells, and vice versa, to directly
compare engraftment and lineage potential between these
two populations in the same recipient animals. We also co-
transplanted 100 adult BM KLS cells as an internal control.
Cells were sorted from e11.5 YS, AGM, FL, and the vitelline/
umbilical (VU) region, which is also known to contain pre-
HSCs and HSCs at this time point (de Bruijn et al., 2000;
Gordon-Keylock et al., 2013). Because e11.5 hematopoietic
progenitors have poor engraftment potential when trans-
planted into adult animals, we instead transplanted intrave-
nously into irradiated newborn nonobese diabetic-severe
combined immunodeficiency-gc
/
(NSG) recipients.
When we examined donor chimerism in the blood of recip-
ient mice, we found substantially more donor cells derived
from CD11A
KLS cells than from CD11A
+
KLS cells in all
recipient mice (Figures 7A and 7B), despite transplanting
nearly a 5-fold excess of CD11A
+
KLS cells (Figure 7A).
Whereas the level of donor chimerism varied considerably
from recipient to recipient, in every case, the contribution
of CD11A
+
KLS cells was either minor or absent, indicating
these cells are much less effective at engraftment than
CD11A
KLS cells. We also examined the lineage distribu-
tion of donor-derived cells (Figures 7C and 7D). Whereas
all recipients contained donor-derived lymphocytes, only
two had significant donor myeloid cells (FL #1 and
VU #1). We focused on the recipient ‘FL #1’ and found
that CD11A
KLS cells produced robust B cells, T cells,
NK cells, granulocytes, and macrophages (Figure 7C).
Conversely, CD11A
+
KLS cells produced only a modest
number of B cells and Tcells, which faded over time (Figures
7C and 7D). Because erythrocytes and platelets do not
express CD45, a marker we used to distinguish donor and
recipient cells, we were unable to determine whether
CD11A
KLS could produce these lineages, but all other
major hematopoietic lineages were detectable and robust
at 15 weeks posttransplantation, suggesting that CD11A
KLS cells are multipotent both in vivo and in vitro.
DISCUSSION
The essential properties of adult HSCs include the capacity
to maintain themselves indefinitely (self-renewal), the
ability to differentiate into all hematopoietic lineages
(multipotency), and the ability to circulate from the blood
to niches in the bone marrow (engraftability). It remains
unclear whether HSCs develop from hematopoietic-
committed precursor cells that contain some, but not all,
three HSC properties or instead whether HSCs arise de
novo from a hematoendothelial precursor. Earlier studies
had suggested that hematopoietic precursors to HSCs
(i.e., pre-HSCs) exist within the fetus and that these pro-
genitors lacked the ability to home to the bone marrow
but could be matured in vitro into bona fide, bone-
marrow-homing HSCs (Gordon-Keylock et al., 2013; Rybt-
sov et al., 2011). We have adopted an alternative strategy to
identify such precursors to HSCs. We hypothesize that HSC
precursors have the property of multipotency, and we
developed a single-cell assay to reductively identify, quan-
tify, and characterize multipotent cells in the early embryo.
Our data indicate that multipotent cells are predominantly
localized to the YS during the stages when definitive, self-
renewing HSC precursors are thought to arise (e9.5–
e10.5). As the surface marker phenotype of multipotent
CD11A
KLS (VE-CAD
+
CD43
+
KIT
+
CD11A
) cells over-
laps with that of pre-HSCs (VE-CAD
+
CD41
+
CD45
or +
),
CD11A
KLS and pre-HSC populations likely contain the
same precursor cells. The ability of CD11A
KLS cells to
engraft in newborn mice and give rise to multiple hemato-
poietic lineages in vivo confirms the multilineage potential
of this population and strongly implicates CD11A
KLS
cells as a critical intermediate population between the
earliest primitive YS progenitors and fully functional HSCs.
It remains unclear whether HSCs arise from both intra-
embryonic (AGM and FL) and extraembryonic (YS and
PL) tissues or if they emerge from a single site and migrate
elsewhere. Our absolute cell number data indicate that the
YS contains by far the most multipotent cells during the
critical stages during which HSCs arise. Pre-HSCs or HSCs
may also emerge de novo from hemogenic endothelium
in the AGM, but our data suggest that the AGM region by
itself does not produce enough multipotent cells to ac-
count for all the multipotent cells in the embryo and that
likely the vast majority come from the extraembryonic
YS. This is consistent with previous work estimating the
absolute number of HSCs during gestation, suggesting
that both the YS and AGM may contribute to the FL HSC
pool (Kumaravelu et al., 2002). Additionally, our previous
work showing direct orthotopic synchronic transplanta-
tion of e8 to e9 YS cells showed that YS-derived cells can
indeed contribute to complete and lifelong adult hemato-
poiesis (Weissman et al., 1978).
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 467
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
A
B
C
D
(legend on next page)
468 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
CD11A
KLS cells express multiple markers of endothe-
lial cells (EPCR, VE-cadherin, and CD34), suggesting that
they are recently derived from the endothelial lineage
and raising the question of whether they retain endothelial
cell potential and contain hemogenic endothelium.
Whereas we did not observe any endothelial cell differenti-
ation from CD11A
KLS cells in our in vitro cultures, we
were also unable to drive robust hematopoietic colony for-
mation from populations that should contain hemogenic
endothelium (VE-CAD
+
CD34
+
SCA-1
+
CD43
CD41
).
Thus, it is possible that the in vitro assay described here is
incapable of revealing hemogenic endothelium. Whereas
the CD11A
KLS population retains surface markers of
hemogenic endothelium, the defining indicator that the
CD11A
KLS population is hematopoietic is that these
cells can engraft upon intravenous transplantation into
neonatal mice and give rise to multiple hematopoietic
lineages.
Many of the surface markers we used to define embryonic
hematopoietic populations have functional roles in cell
adhesion and trafficking. VE-cadherin is an endothelial
adhesion molecule involved in facilitating the adherens
junctions between endothelial cells (Harris and Nelson,
2010). CD11A is part of a leukocyte adhesion complex
(LFA-1) known to play a role in leukocyte extravasation
from the vasculature by binding to ICAMs on endothelial
cells (Hogg et al., 2011). In a related study, we show that
CD11A is upregulated as HSCs lose self-renewal potential
and may be involved in HSC mobilization out of the
bone marrow (J.W.F., M.A.I., Nathaniel B. Fernhoff, J.S.,
and I.L.W., unpublished data). In this study, we show
that CD11A upregulation is also associated with the loss
of multilineage potential and marks a subset of lineage-
committed progenitors that are downstream of embryonic
multipotent cells. These changes in adhesion molecule
expression may play an important role in the migration
of progenitors from their tissues of origin (YS, PL, and
AGM) to secondary sites of hematopoiesis (FL and BM).
For example, the downregulation of VE-cadherin may
allow cells to detach from the endothelium and enter circu-
lation, whereas the upregulation of CD11A may allow cells
to exit circulation and seed new sites.
In this study, we created an assay to look for clonal multi-
lineage potential and used it to identify emerging multipo-
tent hematopoietic progenitors within the embryo. Other
markers we focused on were also critical for distinguishing
multipotent cells from downstream progenitors. Due to
space limitations, we include an in-depth technical discus-
sion of these markers and their use in the Supplemental
Information. Our data suggest that a multipotent, self-
renewing hematopoietic wave arises in the YS blood islands
and appears simultaneously in multiple sites at e9.5. We
confirmed the multipotency of this population in vivo.
We hypothesize that this population represents a critical
intermediate in the origins of definitive hematopoiesis,
and armed with a panel of markers that can identify these
cells with high resolution, we can now begin to dissect the
critical steps in the emergence of and maturation of the
first HSCs in embryonic development.
EXPERIMENTAL PROCEDURES
Antibodies
A detailed list of all antibodies used in this study is shown in
Table S1.
Clonal Multipotency Assay
ODT stroma was cultured as described (Vodyanik and Slukvin,
2007). Briefly, ODT was plated onto gelatin-coated dishes and
cultured in the presence of OP9 media (aMEM [made from powder,
Invitrogen catalog No. 12000-022] in 20% serum [Omega Scientific
FB-11]). ODT was passaged around 1:10 every 5 days. OP9 cells
differentiate rapidly to adipocytes when confluent, so great care
was taken to avoid confluence. For clonal assays, ODT was plated
Figure 7. In Vivo Competitive Comparison of Engraftment and Lineage Potential of CD11A
and CD11A
+
KLS
Newborn NSG mice were transplanted with four embryo equivalents of CD11A
KLS and CD11A
+
KLS cells attained from different tissues at
e11.5 along with 100 adult BM KLS cells. Blood was analyzed at 4, 8, 12, and 15 weeks to examine donor chimerism and lineages produced.
(A) Comparison of donor chimerism between BM KLS (green), CD11A
KLS (red), and CD11A
+
KLS (blue). Stacked graphs show the dis-
tribution of donor cells in the blood at 4, 8, 12, and 15 weeks posttransplant, listed as a percentage of total CD45
+
cells (including those of
the recipient). The tissue of origin for e11.5 KLS cells is indicated above each graph. Note that the scale of the y axis is different in each
graph. The number of CD11A
and CD11A
+
KLS cells for each transplant is shown on the table on the right.
(B) Head-to-head comparison of e11.5 donor cells at 4 weeks. Only donor contributions of e11.5 CD11A
KLS (red) and CD11A
+
KLS (blue)
were compared.
(C) FACS analysis of donor lineages from e11.5 FL KLS cells (‘‘FL #1’’) at 15 weeks posttransplant. The percentage displayed for each lineage
is out of that donor’s total cells. For example, the percentage of NK cells shown for CD11A
KLS cells is out of total donor CD11A
KLS-
derived cells.
(D) Time course of the distribution of donor lineages derived from adult BM KLS (top row), CD11A
+
KLS (middle row), and CD11A
KLS
(bottom row) in the blood of recipient mice at 4, 8, 12, and 15 weeks posttransplant. Percentages shown are out of total CD45
+
cells and
shown for NK cells (orange), T cells (purple), B cells (red), macrophages (green), and granulocytes (blue). The gates used to identify each
lineage are shown in (C). Note that the y axis scale is different with each graph.
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 469
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
the day prior to use onto gelatin-coated 96-well plates (Falcon
3072), around 1,000–2,000 cells per well, in OP9 media. The day
of use, the media was replaced with 100 ml of differentiation media
(described in Vodyanik and Slukvin, 2007), aMEM, 10% serum,
100 mM monothioglycerol (catalog No. M6145; Sigma), 50 mg/ml
ascorbic acid (catalog No. A-0278; Sigma), 100 U/ml penicillin/
100 mg/ml streptomycin (catalog No. 15140; Invitrogen), and 13
GlutaMAX (catalog No. 35050; Invitrogen) and the cytokines
SCF (rmSCF; Peprotech 250-03; 10 ng/ml), TPO (rmTPO; Peprotech
315-14; 10 ng/ml), EPO (rhEPO; Invitrogen PHC2054; 0.5 U/ml),
Flt3L (mFlt3L; Peprotech 250-31L; 10 ng/ml), IL-7 (mIL-7; Pepro-
tech 217-17; 10 ng/ml), and IL-15 (IL15/IL15R complex; Ebio-
science 14-8152; 5 ng/ml) were added. Colonies were visibly
confirmed at 3 days after plating. Around 4 to 5 days after plating,
wells with colonies were fed by adding 100 ml of differentiation
media with IL-7 (10 ng/ml), IL-15 (5 ng/ml), and DOX (1 mg/ml).
All wells with colonies at day 3 were harvested, stained, and
analyzed on a BDFortessa FACS analyzer with a high-throughput
sampler on FACS Diva software (BD Biosciences). Colonies were
scored for presence of erythrocytes (TER119
+
), platelets (CD41
+
),
granulocytes (MAC-1
+
, GR1
+
), NK cells (NK1.1
+
), T cells (CD25
+
,
LY6D
+
), and B cells (CD19
+
, LY6D
+
). LY6D is expressed on devel-
oping thymocytes and B cells and was useful as a second marker
to identify T/B lymphocytes (Inlay et al., 2009). Colonies were
scored as MegE if erythrocytes and/or platelets were detected,
GM if granulocytes were detected, and L if NK, T, and/or B cells
were detected. Colonies with MegE, GM, and L potential were
scored as multipotent.
Mouse Embryo Harvest and Cell Sorting
All animal procedures were approved by the International Animal
Care and Use Committee and the Stanford Administrative Panel
on Laboratory Animal Care. Matings of C57B6 mice were estab-
lished and plugs checked in the mornings. Pregnant females
were harvested in the mornings and embryos dissected immedi-
ately. Vitelline and umbilical vessels were typically harvested
with the yolk sac, except where indicated. For the AGM harvest,
the head, tails, feet, and fetal liver/heart were removed, and the
remaining tissue was listed as AGM. Tissues were dissociated in
10 mg/ml Collagenase Type IV (Invitrogen 17104-019) for approx-
imately 30 min to 1 hr, pipetted up and down and filtered in 70
micron mesh. Tissues were typically stained for 15 min on ice in
staining media. See Table S1 for antibodies used. Cells were
analyzed and/or sorted on a BD FACSAria using FACS Diva soft-
ware. For clone sorting, a 100-micron nozzle was used, and cells
were single sorted on ‘single cell’ mode directly onto 96-well
plates with ODT stroma. The authors highly recommend doing a
single sort (as opposed to a double sort) due to substantial loss of
rare cells upon the second sort. We also recommend avoiding
the use of ACK lysis buffer prior to Ab staining, as it dramatically
reduces the VE-cadherin signal.
In Vivo Transplantation and Analysis
CD45.1
+
CFP
+/
males were crossed with CD45.2
+
females to pro-
duce CD45.1
+
CD45.2
+
embryos, half of which were CFP
+
and half
CFP
. CD11A
KLS and CD11A
+
KLS cells from e11.5 tissues were
sorted and pooled such that CFP
+
CD11A
KLS cells were pooled
with CFP
CD11A
+
KLS cells and vice versa. One hundred adult
BM KLS cells (CD45.2
+
CFP
) were also sorted from the mother
and pooled with the sorted e11.5 KLS populations and then trans-
planted via the superficial facial vein into neonatal (days 1–3) NSG
mice (CD45.1
+
) conditioned with 100 rads irradiation. Mice were
bled at 4, 8, 12, and 15 weeks posttransplantation for analysis.
Erythrocytes were lysed in ACK lysis buffer (150 mM NH
4
Cl,
10 mM KHCO
3
, 0.1 mM EDTA), and the remaining cells were
stained with antibodies and analyzed on a BD FACSAria flow
cytometer.
SUPPLEMENTAL INFORMATION
Supplemental Information includes Supplemental Discussion,
Supplemental Experimental Procedures, seven figures, and one
table and can be found with this article online at http://dx.doi.
org/10.1016/j.stemcr.2014.02.001.
AUTHOR CONTRIBUTIONS
M.A.I., T.S., A.M., and I.L.W. designed experiments. M.A.I., T.S.,
and A.M. performed experiments. J.W.F., I.K.D., and J.S. contrib-
uted unpublished data, efforts, and tools. M.A.I. and T.S. wrote
the manuscript, and I.L.W. edited the manuscript. All experiments
were performed in the laboratory of I.L.W.
ACKNOWLEDGMENTS
The authors wish to thank Adriel Cha, Charles Chan, Nate Fernh-
off, Katharina Seiler, Shah Ali, Bevin Brady, Debashis Sahoo, Kyle
Loh, Seth Karten, Pamela Wenzel, and Roger Pedersen for sharing
reagents and expertise; Libuse Jerabek and Terry Storm for lab man-
agement; Teja Naik and Chrissy Muscat for antibody conjugation;
and Charlene Wang, Aaron McCarty, Humberto Contreas-Trujillo,
and Joel Dollaga for mouse management. The research reported in
this article was supported by the National Institutes of Health (5
T32 AI07290 to M.A.I. and J.W.F.; R01HL058770, R01CA86085,
and U01HL09999 to I.L.W.), the California Institute for Stem
Cell Research (T1-00001 to M.A.I. and J.S.; RT2-02060 to I.L.W.),
the Harvard Stem Cell Institute (to T.S.), the Siebel Stem Cell Insti-
tute and the Thomas and Stacey Siebel Foundation (to I.K.D.), and
the Virginia and D.K. Ludwig Fund for Cancer Research (to I.L.W.).
Received: July 31, 2013
Revised: February 5, 2014
Accepted: February 5, 2014
Published: March 20, 2014
REFERENCES
Akashi, K., Traver, D., Miyamoto, T., and Weissman, I.L. (2000). A
clonogenic common myeloid progenitor that gives rise to all
myeloid lineages. Nature 404, 193–197.
Alvarez-Silva, M., Belo-Diabangouaya, P., Salau
¨
n, J., and Dieterlen-
Lie
`
vre, F. (2003). Mouse placenta is a major hematopoietic organ.
Development 130, 5437–5444.
Balazs, A.B., Fabian, A.J., Esmon, C.T., and Mulligan, R.C. (2006).
Endothelial protein C receptor (CD201) explicitly identifies
470 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
hematopoietic stem cells in murine bone marrow. Blood 107,
2317–2321.
Boisset, J.C., van Cappellen, W., Andrieu-Soler, C., Galjart, N.,
Dzierzak, E., and Robin, C. (2010). In vivo imaging of haemato-
poietic cells emerging from the mouse aortic endothelium. Nature
464, 116–120.
Cumano, A., and Godin, I. (2007). Ontogeny of the hematopoietic
system. Annu. Rev. Immunol. 25, 745–785.
Cumano, A., Dieterlen-Lievre, F., and Godin, I. (1996). Lymphoid
potential, probed before circulation in mouse, is restricted to
caudal intraembryonic splanchnopleura. Cell 86, 907–916.
Cumano, A., Ferraz, J.C., Klaine, M., Di Santo, J.P., and Godin, I.
(2001). Intraembryonic, but not yolk sac hematopoietic precur-
sors, isolated before circulation, provide long-term multilineage
reconstitution. Immunity 15, 477–485.
de Bruijn, M.F., Speck, N.A., Peeters, M.C., and Dzierzak, E. (2000).
Definitive hematopoietic stem cells first develop within the major
arterial regions of the mouse embryo. EMBO J. 19, 2465–2474.
Dzierzak, E., and Medvinsky, A. (1995). Mouse embry onic hemato-
poiesis. Trends Genet. 11, 359–366.
Gekas, C., Dieterlen-Lie
`
vre, F., Orkin, S.H., and Mikkola, H.K.
(2005). The placenta is a niche for hematopoietic stem cells. Dev.
Cell 8, 365–375.
Godin, I.E., Garcia-Porrero, J.A., Coutinho, A., Dieterlen-Lie
`
vre, F.,
and Marcos, M.A. (1993). Para-aortic splanchnopleura from early
mouse embryos contains B1a cell progenitors. Nature 364, 67–70.
Godin, I., Dieterlen-Lie
`
vre, F., and Cumano, A. (1995). Emergence
of multipotent hemopoietic cells in the yolk sac and paraaortic
splanchnopleura in mouse embryos, beginning at 8.5 days postcoi-
tus. Proc. Natl. Acad. Sci. USA 92, 773–777.
Gordon-Keylock, S., Sobiesiak, M., Rybtsov, S., Moore, K., and
Medvinsky, A. (2013). Mouse extraembryonic arterial vessels
harbor precursors capable of maturing into definitive HSCs. Blood
122, 2338–2345.
Harris, E.S., and Nelson, W.J. (2010). VE-cadherin: at the front,
center, and sides of endothelial cell organization and function.
Curr. Opin. Cell Biol. 22, 651–658.
Hibbs, M.L., Xu, H., Stacker, S.A., and Springer, T.A. (1991). Regu-
lation of adhesion of ICAM-1 by the cytoplasmic domain of
LFA-1 integrin beta subunit. Science 251, 1611–1613.
Hogg, N., Patzak, I., and Willenbrock, F. (2011). The insider’s guide
to leukocyte integrin signalling and function. Nat. Rev. Immunol.
11, 416–426.
Huang, H., and Auerbach, R. (1993). Identification and character-
ization of hematopoietic stem cells from the yolk sac of the early
mouse embry o. Proc. Natl. Acad. Sci. USA 90, 10110–10114.
Inlay, M.A., Bhattacharya, D., Sahoo, D., Serwold, T., Seita, J.,
Karsunky, H., Plevritis, S.K., Dill, D.L., and Weissman, I.L. (2009).
Ly6d marks the earliest stage of B-cell specification and identifies
the branchpoint between B-cell and T-cell development. Genes
Dev. 23, 2376–2381.
Iwasaki, H., Arai, F., Kubota, Y., Dahl, M., and Suda, T. (2010). Endo-
thelial protein C receptor-expressing hematopoietic stem cells
reside in the perisinusoidal niche in fetal liver. Blood 116, 544–553.
Karsunky, H., Inlay, M.A., Serwold, T., Bhattacharya, D., and Weiss-
man, I.L. (2008). Flk2+ common lymphoid progenitors possess
equivalent differentiation potential for the B and T lineages. Blood
111, 5562–5570.
Kisanuki, Y.Y., Hammer, R.E., Miyazaki, J., Williams, S.C., Richard-
son, J.A., and Yanagisawa, M. (2001). Tie2-Cre transgenic mice: a
new model for endothelial cell-lineage analysis in vivo. Dev. Biol.
230, 230–242.
Kodama, H., Nose, M., Niida, S., Nishikawa, S., and Nishikawa, S.
(1994). Involvement of the c-kit receptor in the adhesion of
hematopoietic stem cells to stromal cells. Exp. Hematol. 22,
979–984.
Kumaravelu, P., Hook, L., Morrison, A.M., Ure, J., Zhao, S., Zuyev,
S., Ansell, J., and Medvinsky, A. (2002). Quantitative develop-
mental anatomy of definitive haematopoietic stem cells/
long-term repopulating units (HSC/RUs): role of the aorta-gonad-
mesonephros (AGM) region and the yolk sac in colonisation of
the mouse embryonic liver. Development 129, 4891–4899.
Lux, C.T., Yoshimoto, M., McGrath, K., Conway, S.J., Palis, J., and
Yoder, M.C. (2008). All primitive and definitive hematopoietic
progenitor cells emerging before E10 in the mouse embryo are
products of the yolk sac. Blood 111, 3435–3438.
Medvinsky, A., and Dzierzak, E. (1996). Definitive hematopoiesis is
autonomously initiated by the AGM region. Cell 86, 897–906.
Medvinsky, A.L., Samoylina, N.L., Mu
¨
ller, A.M., and Dzierzak, E.A.
(1993). An early pre-liver intraembryonic source of CFU-S in the
developing mouse. Nature 364, 64–67.
Monvoisin, A., Alva, J.A., Hofmann, J.J., Zovein, A.C., Lane, T.F.,
and Iruela-Arispe, M.L. (2006). VE-cadherin-CreERT2 transgenic
mouse: a model for inducible recombination in the endothelium.
Dev. Dyn. 235, 3413–3422.
Moore, M.A., and Metcalf, D. (1970). Ontogeny of the haemo-
poietic system: yolk sac origin of in vivo and in vitro colony form-
ing cells in the developing mouse embryo. Br. J. Haematol. 18,
279–296.
Morrison, S.J., Hemmati, H.D., Wandycz, A.M., and Weissman, I.L.
(1995). The purification and characterization of fetal liver hemato-
poietic stem cells. Proc. Natl. Acad. Sci. USA 92, 10302–10306.
Mu
¨
ller, A.M., Medvinsky, A., Strouboulis, J., Grosveld, F., and
Dzierzak, E. (1994). Development of hematopoietic stem cell activ-
ity in the mouse embryo. Immunity 1, 291–301.
Muzumdar, M.D., Tasic, B., Miyamichi, K., Li, L., and Luo, L.
(2007). A global double-fluorescent Cre reporter mouse. Genesis
45, 593–605.
Nishikawa, S.I., Nishikawa, S., Kawamoto, H., Yoshida, H., Kizu-
moto, M., Kataoka, H., and Katsura, Y. (1998). In vitro generation
of lymphohematopoietic cells from endothelial cells purified
from murine embryos. Immunity 8, 761–769.
Ottersbach, K., and Dzierzak, E. (2005). The murine placenta
contains hematopoietic stem cells within the vascular labyrinth
region. Dev. Cell 8, 377–387.
Palis, J., Robertson, S., Kennedy, M., Wall, C., and Keller, G. (1999).
Development of erythroid and myeloid progenitors in the yolk sac
and embryo proper of the mouse. Development 126, 5073–5084.
Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors 471
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
Rhodes, K.E., Gekas, C., Wang, Y., Lux, C.T., Francis, C.S., Chan,
D.N., Conway, S., Orkin, S.H., Yoder, M.C., and Mikkola, H.K.
(2008). The emergence of hematopoietic stem cells is initiated in
the placental vasculature in the absence of circulation. Cell Stem
Cell 2, 252–263.
Rybtsov, S., Sobiesiak, M., Taoudi, S., Souilhol, C., Senserrich, J.,
Liakhovitskaia, A., Ivanovs, A., Frampton, J., Zhao, S., and Medvin-
sky, A. (2011). Hierarchical organization and early hematopoietic
specification of the developing HSC lineage in the AGM region.
J. Exp. Med. 208, 1305–1315.
Samokhvalov, I.M., Samokhvalova, N.I., and Nishikawa, S. (2007).
Cell tracing shows the contribution of the yolk sac to adult haema-
topoiesis. Nature 446, 1056–1061.
Schmitt, T.M., and Zu
´
n
˜
iga-Pflu
¨
cker, J.C. (2002). Induction of T cell
development from hematopoietic progenitor cells by delta-like-1
in vitro. Immunity 17, 749–756.
Tanaka, Y., Hayashi, M., Kubota, Y., Nagai, H., Sheng, G., Nishi-
kawa, S., and Samokhvalov, I.M. (2012). Early ontogenic origin
of the hematopoietic stem cell lineage. Proc. Natl. Acad. Sci. USA
109, 4515–4520.
Taoudi, S., Gonneau, C., Moore, K., Sheridan, J.M., Blackburn,
C.C., Taylor, E., and Medvinsky, A. (2008). Extensive hematopoiet-
ic stem cell generation in the AGM region via maturation of
VE-cadherin+CD45+ pre-definitive HSCs. Cell Stem Cell 3, 99–108.
Vodyanik, M.A., and Slukvin, I.I. (2007). Hematoendothelial
differentiation of human embryonic stem cells. Curr. Protoc. Cell
Biol. Chapter 23,6.
Weissman, I.L., Baird, S., Gardner, R.L., Papaioannou, V.E., and
Raschke, W. (1977). Normal and neoplastic maturation of T-line-
age lymphocytes. Cold Spring Harb. Symp. Quant. Biol. 41, 9–21.
Weissman, I., Papaioannou, V., and Gardner, R. (1978). Fetal
hematopoietic origin of the adult hematolymphoid system. In
Differentiation of Normal and Neoplastic Hematopoietic Cells, B.
Clarkson, P.A. Marks, and J.E. Till, eds. (Cold Spring Harbor: Cold
Spring Harbor Laboratory Press), pp. 33–47.
Yoder, M.C., and Hiatt, K. (1997). Engraftment of embryonic
hematopoietic cells in conditioned newborn recipients. Blood
89, 2176–2183.
Yoder, M.C., Hiatt, K., Dutt, P., Mukherjee, P., Bodine, D.M., and
Orlic, D. (1997a). Characterization of definitive lymphohemato-
poietic stem cells in the day 9 murine yolk sac. Immunity 7,
335–344.
Yoder, M.C., Hiatt, K., and Mukherjee, P. (1997b). In vivo repopu-
lating hematopoietic stem cells are present in the murine yolk sac
at day 9.0 postcoitus. Proc. Natl. Acad. Sci. USA 94, 6776–6780.
Zovein, A.C., Hofmann, J.J., Lynch, M., French, W.J., Turlo, K.A.,
Yang, Y., Becker, M.S., Zanetta, L., Dejana, E., Gasson, J.C., et al.
(2008). Fate tracing reveals the endothelial origin of hematopoietic
stem cells. Cell Stem Cell 3, 625–636.
472 Stem Cell Reports j Vol. 2 j 457–472 j April 8, 2014 j ª2014 The Authors
Stem Cell Reports
Mouse Embryonic Multipotent Hematopoietic Cells
... There are sequential waves of hematopoiesis, each producing different lineages that increase in their complexity and diversity of blood lineage potential (Costa et al., 2012;Dzierzak and Bigas, 2018). The earliest two waves of hematopoiesis mainly produce primitive hematopoietic cells, lineage-restricted progenitor cells and multipotent progenitor cells (MPPs) in a hematopoietic stem cell (HSC)-independent manner (Dignum et al., 2021;Frame et al., 2013;Inlay et al., 2014;Palis et al., 1999;Patel et al., 2022;Soares-da-Silva et al., 2021). In mice, the primitive hematopoietic cells, such as erythrocytes, megakaryocytes and myeloid cells, are found in the yolk sac blood islands derived from extra-embryonic mesoderm at embryonic day (E) 7.5 and primarily arise from unipotent hematopoietic progenitor cells (HPCs) Palis, 2016;Soares-da-Silva et al., 2021). ...
... For example, in zebrafish, lymphoiderythroid progenitor cells and lymphoid-myeloid progenitor cells, which arise from aortic HE, maintain major developmental hematopoiesis (Ulloa et al., 2021). In mice, an immune-related hematopoietic wave is also detected in the yolk sac, which can produce lymphoid-myeloid progenitor cells and B-1a cells (Boiers et al., 2013;Hadland and Yoshimoto, 2018;Inlay et al., 2014;Yoshimoto et al., 2011). Furthermore, MPPs have also been identified before, or concurrently with, HSC generation; e.g. recently it has been shown that the earliest MPPs emerge at E9-E10 from the para-aortic splanchnopleura/aorta-gonad-mesonephros (AGM) and are derived from Cxcr4-negative, progenitor-restricted HE in mice (Dignum et al., 2021). ...
Article
For a long time, self-renewing and multipotent hematopoietic stem cells (HSCs) have been thought to make a major contribution to both embryonic and adult hematopoiesis. The canonical hematopoietic hierarchy illustrating HSC self-renewal and multipotency has been established mainly based on invasive functional assays (e.g. transplantation or colony-forming units in the spleen and in culture), which evaluate the cellular potentials of HSCs. With the extensive applications of non-invasive cell fate-mapping strategies, recent lineage tracing-based studies have suggested that not all native hematopoiesis is established via the hierarchical differentiation of HSCs. By contrast, hematopoietic progenitor cells (HPCs) are a dominant contributor to both embryonic and young adult hematopoiesis. These new findings help redefine the cellular origins of embryonic and adult hematopoiesis under native conditions, and emphasize the differences in revealing HSC potential versus HSC fate using distinct approaches during stress and native hematopoiesis. Here, we review recent advances in HPC and HSC development, and provide an updated perspective to incorporate these new findings with our traditional understanding of developmental and adult hematopoiesis.
... In mammals, successive waves of hematopoiesis occur at various anatomic sites during embryogenesis (Figure 2a) (Ghosn et al., 2019;Inlay et al., 2014). The initial wave arises in the yolk sac blood islands at embryonic day (E) 7.5 and generates primitive erythrocytes. ...
... This developmental history generates diversity in the hematopoietic potential of HSCs. It has also been shown that yolk sac progenitor cells, which were previously believed to be transient hematopoietic cells during embryogenesis, are involved in adult hematopoiesis (Inlay et al., 2014;Lee et al., 2016;Neo et al., 2021). Although these yolk-sac-derived hematopoietic progenitor cells were previously thought to be lineage restricted, they can differentiate into the mature blood cells of several adult hematopoietic lineages. ...
Article
Full-text available
Tissue stem cells are vital for organ homeostasis and regeneration due to their ability to self-renew and differentiate into the various cell types that constitute organ tissue. These stem cells are formed during complex and dynamic organ development, necessitating spatial-temporal coordination of morphogenetic events and cell fate specification during this process. In recent years, technological advances have enabled the tracing of the cellular dynamics, states, and lineages of individual cells over time in relation to tissue morphological changes. These dynamic data have not only revealed the origin of tissue stem cells in various organs but have also led to an understanding of the molecular, cellular, and biophysical bases of tissue stem cell formation. Herein, we summarize recent findings on the developmental origin of tissue stem cells in the hair follicles, intestines, brain, skeletal muscles, and hematopoietic system, and further discuss how stem cell fate specification is coordinated with tissue topology.
... 58 In addition, intraembryonic hematopoiesis does not exclusively produce HSCs, as nonengrafting multipotent progenitors, as well as bi-and unipotent progenitors derived from the embryo proper before HSC emergence have been described by multiple independent groups. [59][60][61][62] Although researchers have noted that certain T cell subsets are preferentially derived from progenitors that emerge before HSC development, 63 functional differences between the T cells derived from yolk sac progenitors, intraembryonic progenitors, and HSCs have not yet been characterized. ...
Article
Full-text available
Engineered T cells are at the leading edge of clinical cell therapy. T cell therapies have had a remarkable impact on patient care for a subset of hematological malignancies. This foundation has motivated the development of off-the-shelf engineered cell therapies for a broad range of devastating indications. Achieving this vision will require cost-effective manufacturing of precision cell products capable of addressing multiple process and clinical-design challenges. Pluripotent stem cell (PSC)-derived engineered T cells are emerging as a solution of choice. To unleash the full potential of PSC-derived T cell therapies, the field will require technologies capable of robustly orchestrating the complex series of time- and dose-dependent signaling events needed to recreate functional T cell development in the laboratory. In this article, we review the current state of allogenic T cell therapies, focusing on strategies to generate engineered lymphoid cells from PSCs. We highlight exciting recent progress in this field and outline timely opportunities for advancement with an emphasis on niche engineering and synthetic biology.
... 121 Indeed, there is a wave of hematopoietic cells that initiates at ~E9 and is characterized by a sudden rise in multipotent progenitors, including some with a lymphoid bias, known as the lymphoid-primed multipotent progenitors (LMPPs). [122][123][124][125][126] The LMPPs are responsible for the production of some innate lym- There have also been studies showing that susceptibility to autoimmunity peaks during specific windows of development. 132 Thus, future studies are needed to more closely examine how age-related changes in tolerance contribute to autoimmunity in adulthood. ...
Article
Full-text available
Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8⁺ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8⁺ T cells are created equally and that the functions of individual CD8⁺ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8⁺ T cells and propose that the layered immune model be extended to the CD8⁺ T cell compartment.
... Single BM cell suspensions were stained using anti-mouse Abs against B220 (clone RA3-6B2; eBioscience), CD19 (clone 1D3; eBioscience), CD43 (clone S7; BD Biosciences, Franklin Lakes, NJ), IgM (clone RMM-1; BioLegend), IgD (clone IA6-2; BioLegend), c-Kit (clone 2B8; eBioscience), Flk2 (clone A2F10; eBioscience) and Ter119 (clone Ter119; eBioscience). The gating was done as described (17,18). ...
Article
Full-text available
We have identified a novel mouse and human cytokine. IL-40 is normally produced in the bone marrow and fetal liver, and by activated B cells. Its sequence predicts a small (~27kDa) secreted protein unrelated to other cytokine gene families, that we have called Interleukin 40 (IL-40). IL40 is only present in mammalian genomes, suggesting a role in mammalian immune responses. Accordingly, IL-40 expression is induced in the mammary gland upon the onset of lactation, and an IL-40−/− mouse exhibits reduced levels of IgA in the serum, gut, feces, and milk. Furthermore, the IL-40−/− mouse has smaller and fewer Peyer’s patches and IgA secreting cells. The gut microbiome of IL-40−/− mice also exhibits altered composition, reflecting the reduced levels of IgA in the gut. We have also determined that B cell precursor populations are altered in the IL-40−/− mouse. Taken together, these observations indicate that IL-40 represents a novel B cell associated cytokine, that plays an important role in the development of humoral immune responses. IL-40 is also expressed by human activated B cells and by several human B cell lymphomas. The latter observation suggests that it may play a role in the pathogenesis of these diseases.
Article
Hematopoietic stem cells (HSCs) give rise to nearly all blood cell types and play a central role in blood cell production in adulthood. For many years it was assumed that these roles were similarly responsible for driving the formation of the hematopoietic system during the embryonic period. However, detailed analysis of embryonic hematopoiesis has revealed the presence of hematopoietic cells that develop independently of HSCs both before and after HSC generation. Furthermore, it is becoming increasingly clear that HSCs are less involved in the production of functioning blood cells during the embryonic period when there is a much higher contribution from HSC-independent hematopoietic processes. We outline the current understanding and arguments for HSC-dependent and -independent hematopoiesis, mainly focusing on mouse ontogeny.
Article
Full-text available
The adult-definitive hematopoietic hierarchy and hematopoietic stem cells (HSCs) residing in the bone marrow are established during embryonic development. In mouse, human and many other mammals, it is the sudden formation of so-called intra-aortic/arterial hematopoietic clusters (IAHCs) that best signifies and visualizes this de novo generation of HSCs and hematopoietic progenitor cells (HPCs). Cluster cells arise through an endothelial-to-hematopoietic transition and, for some time, express markers/genes of both tissue types, whilst acquiring more hematopoietic features and losing endothelial ones. Amongst several hundreds of IAHC cells, the midgestation mouse embryo contains only very few bona fide adult-repopulating HSCs, suggestive of a challenging cell fate to achieve. Most others are HPCs of various types, some of which have the potential to mature into HSCs in vitro. Based on the number of cells that reveal hematopoietic function, a fraction of IAHC cells is uncharacterized. This review aims to explore the current state of knowledge on IAHC cells. We will describe markers useful for isolation and characterization of these fleetingly-produced yet vitally-important cells and for the refined enrichment of the HSCs they contain, and speculate on the role of some IAHC cells that are as-yet functionally uncharacterized. Twitter Abstract: Here we explore the current state of knowledge on the vitally important intra-aortic hematopoietic cluster cells that arise during development and contain the earliest functional hematopoietic stem cells.
Article
Bone remodeling is a continuous and dynamic process of bone formation and resorption to maintain its integrity and homeostasis. Bone marrow is a source of various cell lineages, including osteoblasts and osteoclasts, which are involved in bone formation and resorption, respectively, to maintain bone homeostasis. Epigenetics is one of the elementary regulations governing the physiology of bone remodeling. Epigenetic modifications, mainly DNA methylation, histone modifications, and non-coding RNAs, regulate stable transcriptional programs without causing specific heritable alterations. DNA methylation in CpG-rich promoters of the gene is primarily correlated with gene silencing, and histone modifications are associated with transcriptional activation/inactivation. However, non-coding RNAs regulate the metastatic potential of cancer cells to metastasize at secondary sites. Deregulated or altered epigenetic modifications are often seen in many cancers and interwound with bone-specific tropism and cancer metastasis. Histone acetyltransferases, histone deacetylase, and DNA methyltransferases are promising targets in epigenetically altered cancer. High throughput epigenome mapping and targeting specific epigenetics modifiers will be helpful in the development of personalized epi-drugs for advanced and bone metastasis cancer patients. This review aims to discuss and gather more knowledge about different epigenetic modifications in bone remodeling and metastasis. Further, it provides new approaches for targeting epigenetic changes and therapy research.
Article
Full-text available
During mouse development, definitive hematopoietic stem cells (dHSCs) emerge by late E10.5 to E11 in several hematopoietic sites. Of them, the aorta-gonad-mesonephros (AGM) region drew particular attention owing to its capacity to autonomously initiate and expand dHSCs in culture, indicating its key role in HSC development. The dorsal aorta contains characteristic hematopoietic clusters and is the initial site of dHSC emergence, where they mature through vascular endothelial (VE)-cadherin(+)CD45(-)CD41(low) (type 1 pre-HSCs) and VE-cadherin(+)CD45(+) (type 2 pre-HSCs) intermediates. Although dHSCs were also found in other embryonic niches (placenta, yolk sac, and extraembryonic vessels), attempts to detect their HSC initiating potential have been unsuccessful to date. Extraembryonic arterial vessels contain hematopoietic clusters, suggesting that they develop HSCs, but functional evidence for this has been lacking. Here we show that umbilical cord and vitelline arteries (VAs), but not veins, contain pre-HSCs capable of maturing into dHSCs in the presence of exogenous interleukin 3, although in fewer numbers than the AGM region, and that pre-HSC activity in VAs increases with proximity to the embryo proper. Our functional data strongly suggest that extraembryonic arteries can actively contribute to adult hematopoiesis.
Article
Full-text available
We show by an in vitro approach that multipotent hemopoietic cells can be detected in the body of the mouse embryo between the stages of 10-25 somites (8.5-9.5 days of gestation)-i.e., prior to liver colonization (28-32 pairs of somites). Interestingly, hemopoietic cells appear in parallel in this location, the paraaortic splanchnopleura, and in the yolk sac, where they represent a new generation by reference to the primitive hemopoietic stem cells. Lymphoid cell clones, which could differentiate into mature B cells, were obtained from yolk sac and paraaortic splanchnopleura cell preparations but not from other tissues of the embryonic body. These B-cell precursors were first detected around the stage of 10 somites; thereafter, their initial minute numbers increased in parallel in the yolk sac and the paraaortic splanchnopleura, suggesting that their emergence in the two sites was simultaneous. By single cell manipulation, we show that these precursors can generate B and T lymphocytes and myeloid cells; these precursors can thus be defined as multipotent hemopoietic cells.
Article
Full-text available
Several lines of evidence suggest that the adult hematopoietic system has multiple developmental origins, but the ontogenic relationship between nascent hematopoietic populations under this scheme is poorly understood. In an alternative theory, the earliest definitive blood precursors arise from a single anatomical location, which constitutes the cellular source for subsequent hematopoietic populations. To deconvolute hematopoietic ontogeny, we designed an embryo-rescue system in which the key hematopoietic factor Runx1 is reactivated in Runx1-null conceptuses at specific developmental stages. Using complementary in vivo and ex vivo approaches, we provide evidence that definitive hematopoiesis and adult-type hematopoietic stem cells originate predominantly in the nascent extraembryonic mesoderm. Our data also suggest that other anatomical sites that have been proposed to be sources of the definitive hematopoietic hierarchy are unlikely to play a substantial role in de novo blood generation.
Article
Full-text available
The aorta-gonad-mesonephros region plays an important role in hematopoietic stem cell (HSC) development during mouse embryogenesis. The vascular endothelial cadherin⁺ CD45⁺ (VE-cad⁺CD45⁺) population contains the major type of immature pre-HSCs capable of developing into long-term repopulating definitive HSCs. In this study, we developed a new coaggregation culture system, which supports maturation of a novel population of CD45-negative (VE-cad⁺CD45⁻CD41⁺) pre-HSCs into definitive HSCs. The appearance of these pre-HSCs precedes development of the VE-cad⁺CD45⁺ pre-HSCs (termed here type I and type II pre-HSCs, respectively), thus establishing a hierarchical directionality in the developing HSC lineage. By labeling the luminal surface of the dorsal aorta, we show that both type I and type II pre-HSCs are distributed broadly within the endothelial and subendothelial aortic layers, in contrast to mature definitive HSCs which localize to the aortic endothelial layer. In agreement with expression of CD41 in pre-HSCs, in vivo CD41-Cre-mediated genetic tagging occurs in embryonic pre-HSCs and persists in all lymphomyeloid lineages of the adult animal.
Article
Full-text available
The activation of leukocyte integrins through diverse receptors results in transformation of the integrin from a bent, resting form to an extended conformation, which has at least two states of ligand-binding activity. This highly regulated activation process is essential for T cell migration and the formation of an immunological synapse. The signalling events that drive integrin activation are complex. Some key players have been well-characterized, but other aspects of the signalling mechanisms involved are still unclear. This Review focuses on the integrin lymphocyte function-associated antigen 1 (LFA1; also known as αLβ2 integrin), which is expressed by T cells, and explores how disparate signalling pathways synergize to regulate LFA1 activity.
Article
The mouse yolk sac has been shown to contain in-vivo colony forming cells capable of producing granulocytic, megakaryocytic and erythroid spleen colonies; in-vitro colony forming cells producing granulocytic and mononuclear-macrophage colonies in agar; and cells capable of repopuiating the lymphoid and myeloid tissue of lethally irradiated hosts. Similar haemopoietic precursor cells were also demonstrated in the blood at the time of initiation of the circulation and in the early embryonic liver. Organ cultures of 7 day embryos with intact yolk sacs, and embryos or yolk sacs after separation have shown the autonomous nature of the development of haemopoiesis in the yolk sac and the dependence of intra-embryonic haemopoiesis, particularly in embryonic liver, on colonization by yolk sac haemopoietic cells. Both in-vivo and in-vitro colony forming cells have been involved in the first migration stream, between yolk sac and embryonic liver, and evidence has been presented for the role of local environmental factors in controlling the differentiation of these cell types. These results support the view that development of haemopoietic organs in both embryo and adult is dependent on colonization by circulating cells and that these circulating stem cells originate initially in the yolk sac. This indicates that the yolk sac is the only site of genuine de novo formation of haemopoietic stem cells.
Article
Endothelial cells form cell-cell adhesive structures, called adherens and tight junctions, which maintain tissue integrity, but must be dynamic for leukocyte transmigration during the inflammatory response and cellular remodeling during angiogenesis. This review will focus on Vascular Endothelial (VE)-cadherin, an endothelial-specific cell-cell adhesion protein of the adherens junction complex. VE-cadherin plays a key role in endothelial barrier function and angiogenesis, and consequently VE-cadherin availability and function are tightly regulated. VE-cadherin also participates directly and indirectly in intracellular signaling pathways that control cell dynamics and cell cycle progression. Here we highlight recent work that has advanced our understanding of multiple regulatory and signaling mechanisms that converge on VE-cadherin and have consequences for endothelial barrier function and angiogenic remodeling.
Article
Hematopoietic stem cells (HSCs) are maintained in specialized niches in adult bone marrow. However, niche and HSC maintenance mechanism in fetal liver (FL) still remains unclear. Here, we investigated the niche and the molecular mechanism of HSC maintenance in mouse FL using HSCs expressing endothelial protein C receptor (EPCR). The antiapoptotic effect of activated protein C (APC) on EPCR(+) HSCs and the expression of protease-activated receptor 1 (Par-1) mRNA in these cells suggested the involvement of the cytoprotective APC/EPCR/Par-1 pathway in HSC maintenance. Immunohistochemistry revealed that EPCR(+) cells were localized adjacent to, or integrated in, the Lyve-1(+) sinusoidal network, where APC and extracellular matrix (ECM) are abundant, suggesting that HSCs in FL were maintained in the APC- and ECM-rich perisinusoidal niche. EPCR(+) HSCs were in a relatively slow cycling state, consistent with their high expression levels of p57 and p18. Furthermore, the long-term reconstitution activity of EPCR(+) HSCs decreased significantly after short culture but not when cocultured with feeder layer of FL-derived Lyve-1(+) cells, which suggests that the maintenance of the self-renewal activity of FL HSCs largely depended on the interaction with the perisinusoidal niche. In conclusion, EPCR(+) HSCs resided in the perisinusoidal niche in mouse FL.
Article
Haematopoietic stem cells (HSCs), responsible for blood production in the adult mouse, are first detected in the dorsal aorta starting at embryonic day 10.5 (E10.5). Immunohistological analysis of fixed embryo sections has revealed the presence of haematopoietic cell clusters attached to the aortic endothelium where HSCs might localize. The origin of HSCs has long been controversial and several candidates of the direct HSC precursors have been proposed (for review see ref. 7), including a specialized endothelial cell population with a haemogenic potential. Such cells have been described both in vitro in the embryonic stem cell (ESC) culture system and retrospectively in vivo by endothelial lineage tracing and conditional deletion experiments. Whether the transition from haemogenic endothelium to HSC actually occurs in the mouse embryonic aorta is still unclear and requires direct and real-time in vivo observation. To address this issue we used time-lapse confocal imaging and a new dissection procedure to visualize the deeply located aorta. Here we show the dynamic de novo emergence of phenotypically defined HSCs (Sca1(+), c-kit(+), CD41(+)) directly from ventral aortic haemogenic endothelial cells.