ArticlePDF Available

Abstract and Figures

The differentiation of hematopoietic progenitors into erythroid or myeloid cell lineages is thought to depend upon relative levels of the transcription factors gata1 and pu.1. While loss-of-function analysis shows that gata1 is necessary for terminal erythroid differentiation, no study has demonstrated that loss of gata1 alters myeloid differentiation during ontogeny. Here we provide in vivo evidence that loss of Gata1, but not Gata2, transforms primitive blood precursors into myeloid cells, resulting in a massive expansion of granulocytic neutrophils and macrophages at the expense of red blood cells. In addition to this fate change, expression of many erythroid genes was found to be differentially dependent on Gata1 alone, on both Gata1 and Gata2, or independent of both Gata factors, suggesting that multiple pathways regulate erythroid gene expression. Our studies establish a transcriptional hierarchy of Gata factor dependence during hematopoiesis and demonstrate that gata1 plays an integral role in directing myelo-erythroid lineage fate decisions during embryogenesis.
Loss of Gata1 Results in Expanded Myelopoiesis (A) pu.1 expression in control and gata1 MO-injected embryos at 14 somites (a, b), 18 somites (c, d), and 24 hpf (e, f). Embryos (a–d) were flat-mounted with anterior (left) and posterior (right). gata1 MO-injected embryos express pu.1 at 18 somites (d, brackets) and 24 hpf (f) while wild-type embryos have downregulated pu.1 transcripts (c, brackets, e) in the ICM precursors. pu.1 is also expressed in anterior myeloid precursors (a–f). gata1 MO-injected embryos have reduced expression of ␤ e1 globin (h) in the ICM at 24 hpf compared to wild-types (g). (B) The number of cells expressing mpo and l-plastin are increased in the vessels of gata1 morphants (b, f, d, h) compared to wild-type embryos (a, e) and gata2 morphants (c, g). g1 / g2 morphants (d, h) have decreased numbers of mpo - and l-plastin -expressing cells compared to the gata1 morphants alone (b, f). (C) Expression of c-myb (a–d) and ikaros (e–h) is maintained in the ICM cells of gata1 , gata2 , and g1 / g2 morphants at 20 somites. pu.1 expression persists in the ICM of gata1 (j) and g1 / g2 (l) morphants, but not gata2 morphants (k) or wild-type embryos (i). Expression of runx1 (m–p) and cebp ␣ (q–t) persists in gata1 (n, r) and g1 / g2 morphants (p, t) but not wild-type embryos (m, q) or gata2 morphants (o, s) at 22 hpf. cebp ␣ is expressed at low levels in some ICM cells and in gut endoderm (q–t). ICM cells expressing mpo at 22 hpf are reduced in gata2 morphants (w) and lost in gata1 (v) and g1 / g2 morphants (x) compared to wild-type embryos (u). Insets (u–x) are of cranial views of the same embryos showing normal mpo -expressing cells in wild-type (u) and gata1 morphants (v) and reduced numbers of mpo -expressing cells in gata2 (w) and g1 / g2 morphants (x).
… 
Erythroid Precursors Are Converted into Myeloid Precursors in the Absence of Gata1 (A) gata1-gfp cells from wild-type (a) and gata1 splice MO-injected (b) embryos at 10 somites (14 hpf) resemble hematopoietic precursors. At 24 hpf, most wild-type gata1-gfp cells resemble proerythroblasts (c, arrowheads), but myeloid cells with indented nuclei are observed. Cells from gata1 splice MO-injected embryos have myeloid features such as vacuoles and indented nuclei (d, arrowheads). Many erythrocytes and proerythroblasts are isolated from 48 hpf wild-type embryos (e, arrowheads) as well as a few myeloid cells. Cells from 48 hpf gata1 splice MO-injected embryos have vacuoles and indented nuclei (f, arrowheads), suggesting that they are myelomonocytes (scale bars are 10 ␮ m). (B) Wild-type (a) and vlt mutant (c) embryos at 22 hpf have similar numbers of TUNEL-positive cells. vlt mutant embryos have an approximate 2-fold increase in apoptosis in their ICM region (d, inset) compared to their wild-type siblings (b, inset) at 28 hpf. (C) Confocal imaging of transverse sections of DAPI-stained (blue) embryos that have undergone double in situ hybridization for ␤ e1 globin (green) and pu.1 (red). Wild-type embryos express ␤ e1 globin and not pu.1 in their ICM cells at 22 hpf (a–c). gata1 MO-injected embryos express both ␤ e1 globin and pu.1 in their ICM cells at 22 hpf (d–f). Some cells coexpress (yellow) both genes (d–f, arrowheads; n, notochord; nt, neural tube; scale bar equals 40 ␮ m).
… 
Content may be subject to copyright.
Developmental Cell, Vol. 8, 109–116, January, 2005, Copyright ©2005 by Elsevier Inc. DOI 10.1016/j.devcel.2004.12.001
Short ArticleLoss of Gata1 but Not Gata2
Converts Erythropoiesis to
Myelopoiesis in Zebrafish Embryos
inhibit each other’s function by physically interfering
with the transcriptional control of their target genes,
suggesting that the levels of each directly determines
cell fate (Graf, 2002). Despite overexpression studies
demonstrating the importance of GATA1 and PU.1 levels
Jenna L. Galloway,
1
Rebecca A. Wingert,
1
Christine Thisse,
2
Bernard Thisse,
2
and Leonard I. Zon
1,
*
1
Stem Cell Program and
Division of Hematology/Oncology
Children’s Hospital Boston in controlling erythroid or myeloid cell fates, no genetic
evidence has shown that loss of these factors altersDana-Farber Cancer Institute
Howard Hughes Medical Institute myeloid versus erythroid lineage decisions during devel-
opment.Harvard Medical School
Boston, Massachusetts 02115 In zebrafish, expression of hematopoietic genes such
as gata1 and gata2 identifies the first primitive erythroid
3
Institut de Ge
´ne
´tique et de Biologie
Mole
´culaire et Cellulaire precursors at the 5-somite stage in the posterior meso-
derm (reviewed in Davidson and Zon, 2004). After 10UMR 7104
CNRS/INSERM/ULP somites, blood precursors express mature erythroid
markers such as embryonic globin. By24 hr postfertiliza-1 rue Laurent Fries, BP10142
CU de Strasbourg tion (hpf), these cells are encircled by endothelial cells
of the trunk axial vein, and together form the intermedi-67404 Illkirch Cedex
France ate cell mass (ICM), the teleost equivalent of the mam-
malian yolk sac blood island. After 24 hpf, the erythro-
blasts enter circulation, where they later mature into
primitive erythrocytes. Zebrafish myelopoiesis is firstSummary
detected by pu.1 expression in the anterior mesoderm
at the 6-somite stage (Lieschke et al., 2002). pu.1 tran-The differentiation of hematopoietic progenitors into
erythroid or myeloid cell lineages is thought to depend scripts are also found in the ICM blood precursors be-
tween 10 and 18 somites, and at 20 somites maturingupon relative levels of the transcription factors gata1
and pu.1. While loss-of-function analysis shows that myeloid cells in the posterior ICM and anterior head
region express myeloperoxidase (mpo), a granulocytegata1 is necessary for terminal erythroid differentia-
tion, no study has demonstrated that loss of gata1 marker, and l-plastin, a macrophage marker (Bennett et
al., 2001).alters myeloid differentiation during ontogeny. Here
we provide in vivo evidence that loss of Gata1, but In this study, we examined the role of gata1 and gata2
in determining blood cell fate during zebrafish hemato-not Gata2, transforms primitive blood precursors into
myeloid cells, resulting in a massive expansion of poiesis. Despite an absence of erythroid cells in Gata1-
deficient embryos (Lyons et al., 2002), hematopoieticgranulocytic neutrophils and macrophages at the ex-
pense of red blood cells. In addition to this fate change, cells present in the ICM of these embryos were found
to differentiate into granulocytes and macrophages. Byexpression of many erythroid genes was found to be
differentially dependent on Gata1 alone, on both Gata1 expression analysis, confocal microscopy, and cell his-
tology, we show ICM blood precursors have convertedand Gata2, or independent of both Gata factors, sug-
gesting that multiple pathways regulate erythroid gene into myeloid cells in the absence of Gata1. In contrast,
loss of Gata2 did not shift differentiation of the ICM cellsexpression. Our studies establish a transcriptional
hierarchy of Gata factor dependence during hemato- to myeloid lineages. Red blood cell genes were found
to require either Gata1 or both Gata1 and Gata2 or werepoiesis and demonstrate that gata1 plays an integral
role in directing myelo-erythroid lineage fate decisions expressed in the absence of both Gata proteins, sug-
gesting that other transcription factors regulate theirduring embryogenesis.
erythroid expression. Furthermore, our results distin-
guish the functions of Gata1 and Gata2 and highlightIntroduction
the importance of Gata1 in promoting erythroid and sup-
pressing myeloid cell fate decisions in zebrafish.The fate of common myelo-erythroid (CMPs) progenitors
to become megakaryocyte-erythroid progenitors or my-
eloid progenitors is thought to be determined by GATA1 Results
and PU.1 (reviewed in Graf, 2002). Overexpression of
GATA1, a zinc finger transcription factor essential for Loss of Gata1 Results in Expanded Myelopoiesis
erythroid differentiation, reprograms myeloid cells to The mutual antagonism between pu.1 and gata1 and
undergo erythroid and megakaryocytic differentiation their coexpression in the ICM led us to ask if gata1
(Pevny et al., 1995; Iwasaki et al., 2003). Forced expres- regulates pu.1 expression during zebrafish blood devel-
sion of PU.1, an Ets family transcription factor, represses opment. We designed two gata1 morpholinos (MOs) and
erythropoiesis and promotes myeloid differentiation in found them to cause anemia at 36 hpf, phenocopying
erythroid cell lines (Yamada et al., 2001). PU.1 and GATA1 the zebrafish gata1 mutant, vlad tepes (vlt), while a
4-base pair mismatch MO did not (Lyons et al., 2002;
Supplemental Table S1 at http://www.developmentalcell.
*Correspondence: zon@enders.tch.harvard.edu
Developmental Cell
110
Figure 1. Loss of Gata1 Results in Expanded Myelopoiesis
(A) pu.1 expression in control and gata1 MO-injected embryos at 14 somites (a, b), 18 somites (c, d), and 24 hpf (e, f). Embryos (a–d) were
flat-mounted with anterior (left) and posterior (right). gata1 MO-injected embryos express pu.1 at 18 somites (d, brackets) and 24 hpf (f) while
wild-type embryos have downregulated pu.1 transcripts (c, brackets, e) in the ICM precursors. pu.1 is also expressed in anterior myeloid
precursors (a–f). gata1 MO-injected embryos have reduced expression of e1 globin (h) in the ICM at 24 hpf compared to wild-types (g).
(B) The number of cells expressing mpo and l-plastin are increased in the vessels of gata1 morphants (b, f, d, h) compared to wild-type
embryos (a, e) and gata2 morphants (c, g). g1/g2 morphants (d, h) have decreased numbers of mpo- and l-plastin-expressing cells compared
to the gata1 morphants alone (b, f).
(C) Expression of c-myb (a–d) and ikaros (e–h) is maintained in the ICM cells of gata1,gata2, and g1/g2 morphants at 20 somites. pu.1
expression persists in the ICM of gata1 (j) and g1/g2 (l) morphants, but not gata2 morphants (k) or wild-type embryos (i). Expression of runx1
(m–p) and cebp(q–t) persists in gata1 (n, r) and g1/g2 morphants (p, t) but not wild-type embryos (m, q) or gata2 morphants (o, s) at 22 hpf.
cebpis expressed at low levels in some ICM cells and in gut endoderm (q–t). ICM cells expressing mpo at 22 hpf are reduced in gata2
morphants (w) and lost in gata1 (v) and g1/g2 morphants (x) compared to wild-type embryos (u). Insets (u–x) are of cranial views of the same
embryos showing normal mpo-expressing cells in wild-type (u) and gata1 morphants (v) and reduced numbers of mpo-expressing cells in
gata2 (w) and g1/g2 morphants (x).
com/cgi/content/full/8/1/109/DC1/). pu.1 expression in gene expression in vitro (Yamada et al., 2001), expres-
sion of the granulocyte marker, mpo, and the macro-the ICM and anterior blood region of gata1 morphants
resembled that of wild-type embryos at 5, 12, and 14 phage marker, l-plastin, were analyzed in Gata1-defi-
cient embryos. Examination of vlt mutants and gata1somites (Figure 1A, a and b; data not shown). Persistent
expression of pu.1 was found in the ICM precursors of morphants at 32 hpf and 4 dpf revealed a significant
increase in the number of mpo (n 36; 100%) andvlt mutants and gata1 morphants from 18 somites to 24
hpf (n 47; 100%; Figure 1A, d and f), whereas in wild- l-plastin (n 37; 100%) positive cells (Figure 1B, b, d,
f, and h; Supplemental Figure S1). These data suggesttype embryos, pu.1 expression was downregulated in
the ICM after 18 somites (Figure 1A, c and e). These data that ICM precursors have differentiated into macro-
phages and neutrophils in the absence of Gata1. Expres-suggest that Gata1 is required to limit pu.1 expression in
ICM precursors after 18 somites. sion of the T cell marker, rag1, was normal in Gata1-
deficient day 4 embryos, demonstrating that Gata1Since forced expression of PU.1 activates myeloid
Gata1 in Zebrafish Myelo-Erythroid Differentiation
111
exclusively regulates myelo-erythroid decisions in ze- Cell Morphology of Gata1-Deficient Cells
To examine Gata1-deficient ICM cell morphology, wild-brafish (n 16; 100%; data not shown).
type and Gata1-deficient ICM cells were isolated from
gata1-gfp transgenic embryos. The gata1-gfp line used
Loss of Gata2 Does Not Cause expresses gfp in the rostral myeloid cells, permitting
Expanded Myelopoiesis both erythroid and myeloid populations to be isolated
To determine if a myeloid expansion occurs in the ab- by flow cytometry (Supplemental Figure S2). GFP-sorted
sence of a related Gata family member also expressed in cells from 14 hpf wild-type and Gata1-deficient embryos
the ICM, a gata2 morpholino and a 4-base pair mismatch morphologically resembled hematopoietic precursors
control morpholino were designed (Supplemental Table (Figure 2A, a and b). GFP cells from wild-type 24 hpf
S1). An increase in mpo- and l-plastin-expressing cells embryos were round or polygonal in shape with coarse
in gata1 morphants at 32 hpf was not observed in gata2 condensed chromatin and a dark blue cytoplasm, char-
morphants (n 30; 0%; Figure 1B, c and g), indicating acteristics that define proerythroblasts (Figure 2A, c). In
the myeloid expansion is specific to loss of gata1.In contrast, an increased number of cells from 24 hpf
mice, loss of GATA2 results in modestly decreased num- Gata1-deficient embryos resembled myeloblasts (wild-
bers of erythroid and myeloid precursors that differenti- type: n 146; 25% myeloblast-like; gata1 MO: n 56;
ate normally (Tsai and Orkin, 1997). Consistent with this, 41% myeloblast-like; Figure 2A, d). At 48 hpf, most wild-
the number of mpo- and l-plastin-expressing myeloid type GFP-sorted cells resembled erythroblasts (Figure
cells at 32 hpf appeared slightly decreased in the gata1/ 2A, e), while many Gata1-deficient cells possessed fea-
gata2 (g1/g2) double morphants (Figure 1B, d and h) tures of promonocytes or maturing granulocytes (wild-
compared to the gata1 morphants alone (Figure 1B, b type: n 363; 13% myeloblast-like; gata1 MO: n
and f). These data suggest that gata1 and gata2 function 210; 43% myeloblast-like; Figure 2A, f, arrowheads). The
distinctly to regulate hematopoietic differentiation and increase in myeloid cells at 48 hpf provides further evi-
cell number, respectively. dence that ICM cells differentiate into myelomonocytes
in the absence of Gata1.
Loss of Gata1 Does Not Affect Expression
of Early Hematopoietic Genes Erythroid Precursors Are Converted into Myeloid
While the transcription factor genes scl,lmo2, and gata2 Precursors in the Absence of Gata1
are expressed in hematopoietic stem cells at 5 somites, As loss of GATA1 in mice causes erythroid progenitors
transcripts of c-myb,runx1, and ikaros are found in both to undergo cell death (Pevny et al., 1995), apoptosis was
primitive erythroid and myeloid progenitors (Davidson examined in zebrafish gata1 mutants by TUNEL staining.
and Zon, 2004). To determine if loss of Gata factors No appreciable increase in cell death was observed in
affects the expression of these genes, in situ hybridiza- vlt mutants (n 6) between 12 somites and 24 hpf
tions were performed with the gata morphants. There (Figure 2B, a and c; data not shown; N. Paffett-Lugassy,
was no significant change in gata2,scl, and lmo2 ex- personal communication). Between 26 and 30 hpf, an
pression at 12 somites (data not shown) and c-myb (n approximate 2-fold increase in TUNEL-positive cells
41; 100%) and ikaros (n 33; 100%; Figure 1C, a–h) was observed in the ICM region of vlt mutants (n 17)
expression at 20 somites in gata1,gata2, and g1/g2 compared to wild-type and heterozygous siblings (n
morphants, indicating that loss of Gata factors does not 20; Figure 2B, b and d). This modest increase in cell
affect early blood formation. death detected in vlt mutants was not significant enough
In wild-type embryos, runx1 is expressed early in ante- to explain the loss of all ICM erythroid precursors.
rior myeloid precursors and ICM cells until 18 somites. To determine if ICM erythroid precursors are con-
In Gata1-deficient embryos, runx1 expression persisted verting into myeloid cells, double in situ hybridizations
in the ICM until 24 hpf (n 37; 100%; Figure 1C, m–p), for e1 globin and pu.1 expression were performed on
suggesting that ICM cells have retained progenitor char- wild-type and Gata1-deficient 22 hpf embryos and ex-
acteristics. Increased expression of CCAAT/enhancer amined by confocal microscopy. In wild-type embryos,
binding protein (cebp), a transcription factor found globin was expressed in ICM cells, while pu.1 was pre-
in myeloid progenitors essential for granulocytic matura- dominantly expressed in the anterior blood region (data
tion (Zhang et al., 1997), was also observed in Gata1- not shown; Figure 2C, a–c). Although pu.1 can be ex-
deficient embryos at 22 hpf (n 24; 100%; Figure 1C, pressed in a few cells in the posterior ICM at 22 hpf,
q–t). Examination of mpo expression prior to circulation wild-type cells were never found to coexpress globin
revealed normal expression in the head region but a lack and pu.1 at this stage. In contrast, multiple ICM cells in
of mpo-expressing cells in the ICM of Gata1-deficient the gata1 morphants were found to coexpress pu.1 and
embryos at 24 hpf (n 46; 100%; Figure 1C, u–x). While globin, suggesting that ICM cells were converting to the
blood cells are specified in Gata1-deficient embryos, an myeloid lineage (Figure 2C, d–f).
absence of mpo-expressing cells at 24 hpf and persis-
tent runx1 expression suggests that the ICM cells are
temporarily delayed in their differentiation. In contrast, A Hierarchy of Gata-Dependent Erythroid
Gene Expressiongata2 morphants never displayed persistent expression
of pu.1 (n 15; 0%), runx1 (n 49; 0%), and cebpTo understand the functional differences between gata1
and gata2 in hematopoietic regulation, expression of(n 15; 0%) after 18 somites, indicating that the myeloid
expansion is specific to loss of Gata1. erythroid genes obtained from an in situ hybridization
Developmental Cell
112
Figure 2. Erythroid Precursors Are Converted into Myeloid Precursors in the Absence of Gata1
(A) gata1-gfp cells from wild-type (a) and gata1 splice MO-injected (b) embryos at 10 somites (14 hpf) resemble hematopoietic precursors.
At 24 hpf, most wild-type gata1-gfp cells resemble proerythroblasts (c, arrowheads), but myeloid cells with indented nuclei are observed.
Cells from gata1 splice MO-injected embryos have myeloid features such as vacuoles and indented nuclei (d, arrowheads). Many erythrocytes
and proerythroblasts are isolated from 48 hpf wild-type embryos (e, arrowheads) as well as a few myeloid cells. Cells from 48 hpf gata1 splice
MO-injected embryos have vacuoles and indented nuclei (f, arrowheads), suggesting that they are myelomonocytes (scale bars are 10 m).
(B) Wild-type (a) and vlt mutant (c) embryos at 22 hpf have similar numbers of TUNEL-positive cells. vlt mutant embryos have an approximate
2-fold increase in apoptosis in their ICM region (d, inset) compared to their wild-type siblings (b, inset) at 28 hpf.
(C) Confocal imaging of transverse sections of DAPI-stained (blue) embryos that have undergone double in situ hybridization for e1 globin
(green) and pu.1 (red). Wild-type embryos express e1 globin and not pu.1 in their ICM cells at 22 hpf (a–c). gata1 MO-injected embryos
express both e1 globin and pu.1 in their ICM cells at 22 hpf (d–f). Some cells coexpress (yellow) both genes (d–f, arrowheads; n, notochord;
nt, neural tube; scale bar equals 40 m).
screen was examined in gata1,gata2, and g1/g2 MO- six genes were unaffected by loss of Gata2, demonstra-
ting that their expression is specifically dependent oninjected embryos (screen described in Supplemental
Experimental Procedures). At 18 somites, epsin (in- gata1.
ICM expression of another group of erythroid genesvolved in clathrin-mediated endocytosis) and GTP bind-
ing-protein 1 (gtpbp1) were normally expressed in ICM was reduced in Gata1-deficient embryos. A decrease in
e1 globin (n 33; 100%), alas2 (n 27; 100%), car-cells; however, in Gata1-deficient embryos, their expres-
sion was absent (Figure 3A, a–d). Expression of biliverdin bonic anhydrase (n 39; 100%), and gata1 (n 33;
100%) expression in gata1 morphants and vlt mutantsreductase (heme degradation), SH3 domain binding pro-
tein 5 (SH3BP5),HIF1-like gene (hypoxic response), was observed (Figure 3B, b, f, j, and n; data not shown).
Previous work in vlt and mouse GATA1-deficient celland 5nucleotidase type B (cause of human hemolytic
anemia; Bianchi et al., 2003) were absent in the ICM of lines also found multiple red cell-specific genes ex-
pressed in the absence of gata1 (Weiss et al., 1994;Gata1-deficient embryos at 20–24 hpf (Figure 3A, e–h;
data not shown). The absence of blood gene expression Lyons et al., 2002). Similar to the reduction in myeloid
cell number, gata2 morphants had a subtle decrease inin Gata1-deficient embryos is consistent with mamma-
lian studies indicating that GATA1 regulates virtually all erythroid cell number as detected by globin (n 16;
100%), alas2 (n 13; 100%), carbonic anhydrase (n red blood cell genes (Orkin, 1992). Furthermore, these
Gata1 in Zebrafish Myelo-Erythroid Differentiation
113
Figure 3. Erythroid Genes Are Differentially Regulated by Gata1 and Gata2
(A) Embryos (a–d) have been flat-mounted and photographed to show only the posterior tail region (anterior, left; posterior, right). Normal
expression of gdpbp1 and epsin (a, c) is lost in ICM erythroid precursors of vlt mutants at 18 hpf (b, d). Expression of biliverdin reductase
and SH3BP5 at 20 hpf (e) and 22 hpf (g), respectively, is absent in vlt mutant ICM cells (f, h).
(B) In situ hybridization for e1 globin (a–d), alas2 (e–h), carbonic anhydrase (i–l), and gata1 (m–p) at 20 hpf. Compared to wild-type embryos
(a, e, i, m), ICM expression of e1 globin,alas2,carbonic anhydrase, and gata1 was decreased in gata1 morphants (b, f, j, n) and absent in
g1/g2 morphants (d, h, l, p). Decreased number of cells expressing e1 globin,alas2,carbonic anhydrase, and gata1 are observed in gata2
morphants (c, g, k, o) compared to wild-types (a, e, i, m).
29; 100%), and gata1 (n 10; 100%) expression at 20 KIAA0650 (n 45; 100%), testhymin (n 45; 100%),
and kelch repeat-containing protein (n 54; 100%) weresomites (Figure 3B, c, g, k, and o). In contrast, globin
(n 118; 91%), alas2 (n 16; 100%), carbonic anhy- expressed in the ICM precursors of the gata1,gata2,
and g1/g2 double morphants at 12 somites (Figure 4A,drase (n 44; 100%), and gata1 (n 18; 100%) expres-
sion was lost in the g1/g2 double morphants compared e–o). We also found that biklf, a Kruppel-like transcrip-
tion factor expressed in early erythroid precursorsto wild-type embryos and gata1 or gata2 single mor-
phants (Figure 3B, d, h, l, and p). This absence of expres- (Oates et al., 2001), was expressed normally in gata1
(n 21; 100%), gata2 (n 16; 100%), and g1/g2 (n sion may result from the additive decrease in erythroid
gene expression, resulting from loss of Gata1 and Gata2. 16; 100%) morphants at 12 somites (Figure 4A, a–d).
The Gata-independent expression of biklf,testhymin,Alternatively, a decline in cell number caused by loss
of Gata2 combined with reduced erythroid gene expres- KIAA0650, and kelch repeat-containing protein sug-
gests that other hematopoietic transcription factors in-sion due to loss of Gata1 may lead to the absence of
expression. Congruent with the former conclusion, sug- duce their expression and that at least part of the ery-
throid program was initiated in gata1 morphants. Aftergesting Gata2 may regulate erythroid gene expression
in the absence of Gata1, mouse studies have found that 20 hpf, gata1 and g1/g2 morphants weakly expressed
biklf (n 87; 100%), KIAA0650 (n 20; 100%), andother GATA factors placed under GATA1 transcriptional
control can substitute for loss of GATA1 during embry- testhymin (n 50; 100%) in the ICM compared to wild-
types or gata2 morphants (Figure 4B), suggesting thatonic hematopoiesis (Takahashi et al., 2000). Neverthe-
less, our results suggest that erythroid genes differ in Gata1 is required to maintainwild-type expression levels
of these genes. Additionally, their downregulation istheir requirements for Gata factors.
concomitant with the conversion of the ICM cells to the
myeloid lineage.Gata-Independent Erythroid Gene Expression
To determine whether loss of both Gata factors ablated
erythroid precursors, we utilized erythroid-specific Discussion
genes identified from the in situ hybridization screen.
The genes KIAA0650,testhymin, and kelch repeat-con- Hierarchy of Regulation of Erythroid
Gene Expressiontaining protein were never detected in the anterior
myeloid cells and were found to be lost in the ICM of 18- As gata1 is a major regulator of erythroid gene expres-
sion, we found that many erythroid genes were depen-somite stage moonshine (mon) embryos, which harbor a
mutation in TIF1-and lack primitive erythrocytes (Ran- dent on gata1 for expression. A subset of erythroid
genes had reduced, but notabsent, expression in Gata1-som et al., 2004; data not shown). This suggests that
these genes are specific to early ICM erythroid cells. deficient embryos. Similarly, the GATA1 null mouse and
Developmental Cell
114
Figure 4. Expression of Gata-Independent Erythroid Genes
(A) Embryos were flat-mounted (anterior, left; posterior, right). Expression of biklf (a–d), KIAA0650 (e–h), testhymin (i–l), and kelch repeat-
containing protein (m–p) is maintained in ICM precursors of 12 somite gata1,gata2, and g1/g2 morphants. Anterior expression of biklf stains
the hatching gland (a–d).
(B) ICM expression of biklf (a–d) at 20 hpf and KIAA0650 (e–h) and testhymin (i–l) at 22 hpf was decreased but present in gata1 morphants
(b, f, j) and g1/g2 MO-injected embryos (d, h, l) compared to wild-type embryos (a, e, i). gata2 morphants (c, g, k) had decreased numbers
of ICM cells expressing biklf,KIAA0650, and testhymin.
GATA1-deficient cell lines exhibited residual expression KIAA0650 (erythroid progenitors). Although attempts to
rescue erythropoiesis in Gata1-deficient embryos byof erythroid genes (Weiss et al., 1994; Pevny et al., 1995).
Our data showing loss of expression of these genes in gata2 overexpression were technically confounded by
gata2 mRNA toxicity (data not shown), studies in mouseG1/G2-deficient embryos suggests that in the absence
of Gata1, Gata2 regulates expression of a subset of have demonstrated that GATA2, when placed under the
control of the GATA1 promoter, can substitute forerythroid genes in erythroid precursors. In support of
this, zebrafish g1/g2 morphants, unlike mouse G1/G2 GATA1 during embryonic hematopoiesis (Takahashi et
al., 2000). This is consistent with the hypothesis thatnulls in which yolk sac blood cells undergo apoptosis
(Fujiwara et al., 2003), have primitive blood cells ex- gata2 regulates some erythroid target genes in the ab-
sence of Gata1.pressing c-myb and ikaros (common progenitors) and
biklf,kelch repeat-containing protein,testhymin, and We discovered that the novel genes kelch repeat-
Gata1 in Zebrafish Myelo-Erythroid Differentiation
115
In Situ Hybridizations and TUNEL
containing protein,testhymin, and KIAA0650 were ex-
Antisense mRNA probes were made for c-myb,biklf,l-plastin,mpo,
pressed independently of Gata factors. In contrast to
pu.1,e1 globin, and gata1 (Thompson et al., 1998; Bennett et al.,
genes like c-myb and ikaros that are found in G1/G2-
2001; Oates et al., 2001; Lieschke et al., 2002). Riboprobes of genes
deficient CMPs, these genes had erythroid-specific ex-
isolated from the in situ hybridization screen were described in
pression, suggesting that they exclusively mark the ery-
Supplemental Experimental Procedures. Confocal imaging was
throid progenitor cell compartment. The early blood
done on paraffin-embedded 10 m sections that underwent double
in situ hybridization (Brent et al., 2003) with Tyramide Signal Amplifi-
genes biklf,scl, and lmo2 may regulate expression of
cation (Perkin Elmer, Molecular Probes). TUNEL was performed us-
these Gata-independent genes, and future studies will
ing the ApopTag Peroxidase In Situ Apoptosis detection kit
further elucidate erythroid gene regulation during hema-
(Chemicon).
topoiesis.
Gene Knockdown by Morpholinos
Gata1 but Not Gata2 Regulates Myelo-Erythroid
Morpholino oligos were designed against the ATG of gata1 (5-CTG
Fate Decisions
CAAGTGTAGTATTGAAGATGTC-3), the first exon/intron boundary of
Our findings demonstrate that gata1 is necessary to
gata1 (5-GTTTGGACTCACCTGGACTGTGTCT-3), and the third
exon/intron boundary of gata2 (5-CATCTACTCACCAGTCTGCGC
promote erythroid cell fate and repress myeloid differen-
TTTG-3). Control morpholinos containing four base pair mismatches
tiation. Similar to vlt, GATA1-deficient mice lack mature
were also designed against the gata1 splice MO (5-GTTCGGACT
red blood cells, and in vitro differentiation of GATA1
CGCCTGTACTGTGTAT-3) and the gata2 splice MO (5-CATCCAC
murine ES cells reveals an increase (5-fold) in myeloid
TCACTAGTCTACGCTGTG-3). Morpholinos were resuspended in
colonies (S. Orkin, personal communication). Despite
nuclease-free water, and one nanoliter was injected at the 1- to
these similarities, an increase in myeloid cells was not
4-cell stage at the following concentrations: 1 mM gata1 MO, 0.2
mM gata2 splice MO, 0.4 mM gata1 splice MO, 0.2 mM gata2 splice
observed in GATA1
and GATA1/GATA2
/
mice and
mismatch MO, and 0.4 mM gata1 splice mismatch MO.
GATA1cultured yolk sac cells (Fujiwara et al., 1996,
2003; Pevny et al., 1995), suggesting that the mouse yolk
Flow Cytometry and Histological Analysis
sac environment may not support this cell fate alteration.
Approximately 200 to 400 uninjected or gata1 splice MO-injected
Loss of zebrafish gata2 does not alter red blood cell
gata1-gfp transgenic embryos were collected (14 hpf, 24 hpf, and
maturation or myeloid differentiation, but does decrease
48 hpf). Embryos were processed, isolated by GFP fluorescence
blood cell number, consistent with decreased numbers
using a FACSVantage flow cytometer (Beckton Dickinson), centri-
of progenitors in GATA2-deficient murine embryonic
fuged, and stained with May-Gru
¨nwald and Giemsa solutions as
described (Traver et al., 2003).
stem cells (Tsai and Orkin, 1997).
Two proposed models may explain the dramatic shift
Acknowledgments
toward myelopoiesis in Gata1-deficient embryos (Graf,
2002). In one model, the ICM contains both myeloid
We would especially like to thank A. Davidson and J. Rivera-Felici-
and erythroid progenitors; in the absence of Gata1, the
ano for critical reading and helpful advice, and acknowledge H.
erythroid progenitors undergo cell death while the my-
Stern, D. Traver, and C. Burns for critical reading, K. Dooley and D.
eloid progenitors expand and differentiate. In the sec-
Ransom for reagents, N. Paffett-Lugassy and S. Orkin for use of
ond model, ICM CMPs differentiate into erythroid or
unpublished data, S. Cho for assistance with confocal imaging, and
A. Flint for assistance with flow cytometry. L.I.Z. is supported by
myeloid cells, based on relative levels of gata1 and pu.1.
HHMI and the NIH (U01DK063328-01). B.T. and C.T. are supported
Our results support the latter model wherein the pres-
by INSERM, CNRS, the Ho
ˆpital Universitaire de Strasbourg, the
ence or absence of Gata1 determines the fate of ICM
Association pour la Recherche sur le Cancer, the Ligue Nationale
progenitors. In Gata1-deficient embryos, blood progeni-
Contre le Cancer, and the NIH (R01RR15402).
tors form normally as demonstrated by expression of
biklf,scl, and lmo2. Without Gata1 antagonism, pu.1
Received: April 19, 2004
expression is not downregulated after 18 somites. This
Revised: September 8, 2004
persistent pu.1 expression is likely the instructive signal
Accepted: December 1, 2004
that guides ICM cells to activate a myeloid differentiation
Published: January 3, 2005
program (Rhodes et al., 2004 [this issue of Develop-
mental Cell]). The decrease in cells expressing biklf and
References
the novel erythroid-specific genes and the presence of
Bennett, C.M., Kanki, J.P., Rhodes, J., Liu, T.X., Paw, B.H., Kieran,
ICM cells coexpressing globin and pu.1 provides strong
M.W., Langenau, D.M., Delahaye-Brown, A., Zon, L.I., Fleming, M.D.,
evidence for an ICM cell fate conversion when Gata1
and Look, A.T. (2001). Myelopoiesis in the zebrafish, Danio rerio.
is absent. Our study demonstrates that gata1 has an
Blood 98, 643–651.
important role in suppressing myeloid differentiation,
Bianchi, P., Fermo, E., Alfinito, F., Vercellati, C., Baserga, M., Ferraro,
presumably by antagonizing the effect on pu.1-initiated
F., Guzzo, I., Rotoli, B., and Zanella, A. (2003). Molecular character-
myelopoiesis. This work also provides genetic evidence
ization of six unrelated Italian patients affected by pyrimidine 5-
that a hierarchy exists in the regulation of erythroid
nucleotidase deficiency. Br. J. Haematol. 122, 847–851.
genes and that gata1, but not gata2, is critical for de-
Brent, A.E., Schweitzer, R., and Tabin, C.J. (2003). A somatic com-
termining erythroid versus myeloid cell fates during em-
partment of tendon progenitors. Cell 113, 235–248.
bryonic development.
Davidson, A., and Zon, L.I. (2004). The ‘definitive’ (and ‘primitive’)
guide to zebrafish hematopoiesis. Oncogene 23, 7233–7246.
Experimental Procedures Fujiwara, Y., Browne, C.P., Cunniff, K., Goff, S.C., and Orkin, S.H.
(1996). Arrested development of embryonic red cell precursors in
Zebrafish Strains mouse embryos lacking transcription factor GATA-1. Proc. Natl.
Zebrafish breeding and staging were done as described (West- Acad. Sci. USA 93, 12355–12358.
erfield, 1994). vlt
m651
were genotyped (Lyons et al., 2002), and the
gata1-gfp line (Long et al., 1997) and wild-type Tu
¨were used. Fujiwara, Y., Chang, A.N., Williams, A.M., and Orkin, S.H. (2003).
Developmental Cell
116
Functional overlap of GATA-1 and GATA-2 in primitive hematopoi-
etic development. Blood 103, 583–585.
Graf, T. (2002). Differentiation plasticity of hematopoietic cells.
Blood 99, 3089–3101.
Iwasaki, H., Mizuno, S., Wells, R.A., Cantor, A.B., Watanabe, S., and
Akashi, K. (2003). GATA-1 converts lymphoid and myelomonocytic
progenitors into the megakaryocyte/erythrocyte lineages. Immunity
19, 451–462.
Lieschke, G.J., Oates, A.C., Paw, B.H., Thompson, M.A., Hall, N.E.,
Ward, A.C., Ho, R.K., Zon, L.I., and Layton, J.E. (2002). Zebrafish
SPI-1 (PU.1) marks a site of myeloid development independent of
primitive erythropoiesis: implications for axial patterning. Dev. Biol.
246, 274–295.
Long, Q., Meng, A., Wang, H., Jessen, J.R., Farrell, M.J., and Lin,
S. (1997). GATA-1 expression pattern can be recapitulated in living
transgenic zebrafish using GFP reporter gene. Development 124,
4105–4111.
Lyons, S.E., Lawson, N.D., Lei, L., Bennett, P.E., Weinstein, B.M.,
and Liu, P.P. (2002). A nonsense mutation in zebrafish gata1 causes
the bloodless phenotype in vlad tepes. Proc. Natl. Acad. Sci. USA
99, 5454–5459.
Oates, A.C., Pratt, S.J., Vail, B., Yan, Y.l., Ho, R.K., Johnson, S.L.,
Postlethwait, J.H., and Zon, L.I. (2001). The zebrafish klf gene family.
Blood 98, 1792–1801.
Orkin, S.H. (1992). GATA-binding transcription factors in hematopoi-
etic cells. Blood 80, 575–581.
Pevny, L., Lin, C., D’Agati, V., Simon, M.C., Orkin, S.H., and Costan-
tini, F. (1995). Development of hematopoietic cells lacking transcrip-
tion factor GATA-1. Development 121, 163–172.
Ransom, D.G., Bahary, N., Niss, K., Traver, D., Burns, C., Paffett-
Lugassy, N., Saganic, W.J., Lim, C.A., Hersey, C., Zhou, Y., et al.
(2004). The zebrafish moonshine gene encodes transcriptional inter-
mediary factor 1, an essential regulator of hematopoiesis. PLoS
2, 1188–1196.
Rhodes, J., Hagen, A., Hsu, K., Deng, M., Xi Liu, T., Look, A.T., and
Kanki, J.P. (2004). Interplay of Pu.1 and Gata1 determines myelo-
erythroid progenitor cell fate in zebrafish. Dev. Cell 8, this issue,
97–108.
Takahashi, S., Shimizu, R., Suwabe, N., Kuroha, T., Yoh, K., Ohta,
J., Nishimura, S., Lim, K.C., Engel, J.D., and Yamamoto, M. (2000).
GATA factor transgenes under GATA-1 locus control rescue germ-
line GATA-1 mutant deficiencies. Blood 96, 910–916.
Thompson, M.A., Ransom, D.G., Pratt, S.J., MacLennan, H., Kieran,
M.W., Detrich, H.W., Vail, B., Huber, T.L., Paw, B., Brownlie, B.,
et al. (1998). The cloche and spadetail genes differentially affect
hematopoiesis and vasculogenesis. Dev. Biol. 197, 248–269.
Traver, D., Paw, B.H., Poss, K.D., Penberthy, W.T., Lin, S., and Zon,
L.I. (2003). Transplantation and in vivo imaging of multilineage en-
graftment in zebrafish bloodless mutants. Nat. Immunol. 4, 1238–
1244.
Tsai, F.Y., and Orkin, S.H. (1997). Transcription factor GATA-2 is
required for proliferation/survival of early hematopoietic cells and
mast cell formation, but not for erythroid and myeloid terminal differ-
entiation. Blood 89, 3636–3643.
Weiss, M.J., Keller, G., and Orkin, S.H. (1994). Novel insights into
erythroid development revealed through in vitro differentiation of
GATA-1 embryonic stem cells. Genes Dev. 8, 1184–1197.
Westerfield, M. (1994). The Zebrafish Book: A Guide for the Labora-
tory Use of Zebrafish (Brachydanio rerio), 2.1 Edition (Eugene, OR:
University of Oregon Press).
Yamada, T., Abe, M., Higashi, T., Yamamoto, H., Kihara-Negishi, F.,
Sakurai, T., Shirai, T., and Oikawa, T. (2001). Lineage switch induced
by overexpression of Ets family transcription factor PU.1 in murine
erythroleukemia cells. Blood 97, 2300–2307.
Zhang, D.E., Zhang, P., Wang, N.D., Hetherington, C.J., Darlington,
G.J., and Tenen, D.G. (1997). Absence of granulocyte colony-stimu-
lating factor signaling and neutrophil development in CCAAT en-
hancer binding protein alpha-deficient mice. Proc. Natl. Acad. Sci.
USA 94, 569–574.
... Scale bar = 500 μm. Mean ± SEM. erythrocytes [66][67][68][69][70][71]. Hence, we decided to use the Tg(gata1:ds-Red) sd2 transgenic line [67], in which erythroid cells display a strong red fluorescence, to see whether Stat3 has a role in hypoxia-induced erythropoiesis. ...
... Mean ± SEM. erythrocytes [66][67][68][69][70][71]. Hence, we decided to use the Tg(gata1:ds-Red) sd2 transgenic line [67], in which erythroid cells display a strong red fluorescence, to see whether Stat3 has a role in hypoxia-induced erythropoiesis. Notably, 6 h long hypoxia treatments determine a significant increase of fluorescence in stat3 +/+ (Fig. 5C, D). ...
... stat3 ia23 mutant line [44] is genotyped by PCR amplification and 3% agarose gel migration. The Tg(7xStat3-Hsv.Ul23:EGFP) ia28 line, the Tg(4xHRE-TATA:mCherry,cmlc2:EGFP) ia22 line, the Tg(fli1:EGFP) y1 , the Tg(gata1:dsRed) sd2 line and the Tg(LysC:dsRed) nz50 lines have been respectively characterized by Peron et al. [44], Vettori et al. [39], Lawson and Weinstein [60], Galloway et al. [67] and Hall et al. [76]. All animal experiments were performed under the permission of the ethical committee of the University of Padova and the Italian Ministero della Salute (23/2015-PR). ...
Article
Full-text available
STAT3 and HIF1α are two fundamental transcription factors involved in many merging processes, like angiogenesis, metabolism, and cell differentiation. Notably, under pathological conditions, the two factors have been shown to interact genetically, but both the molecular mechanisms underlying such interactions and their relevance under physiological conditions remain unclear. In mouse embryonic stem cells (ESCs) we manage to determine the specific subset of hypoxia-induced genes that need STAT3 to be properly transcribed and, among them, fundamental genes like Vegfa , Hk1 , Hk2 , Pfkp and Hilpda are worth mentioning. Unexpectedly, we also demonstrated that the absence of STAT3 does not affect the expression of Hif1α mRNA nor the stabilization of HIF1 α protein, but the STAT3-driven regulation of the hypoxia-dependent subset of gene could rely on the physical interaction between STAT3 and HIF1 α . To further elucidate the physiological roles of this STAT3 non-canonical nuclear activity, we used a CRISPR/Cas9 zebrafish stat3 knock-out line. Notably, hypoxia-related fluorescence of the hypoxia zebrafish reporter line (HRE:mCherry) cannot be induced when Stat3 is not active and, while Stat3 Y705 phosphorylation seems to have a pivotal role in this process, S727 does not affect the Stat3-dependent hypoxia response. Hypoxia is fundamental for vascularization, angiogenesis and immune cells mobilization; all processes that, surprisingly, cannot be induced by low oxygen levels when Stat3 is genetically ablated. All in all, here we report the specific STAT3/HIF1 α -dependent subset of genes in vitro and, for the first time with an in vivo model, we determined some of the physiological roles of STAT3-hypoxia crosstalk.
... In addition, we examined the phenotypes of brain pericytes in the gata1a morphants, which exhibited loss of blood red cells and reduced hemodynamic effect on blood vessels. 38,39 In comparison with the standard morpholino (MO)-injected controls, gata1a morphants showed a significant reduction in both the brain pericyte number and the pericyte coverage of brain vessels (number: gata1a MO = 55.8 ± 2.5 versus std MO = 70.6 ± 1.6, decreased by 21%, p < 0.001; coverage: gata1a MO = 0.31 ± 0.01 versus std MO = 0.45 ± 0.01, decreased by 31%, p < 0.0001) ( Figures S3H-S3J). These results, together with the data of pharmacological interventions, provide evidence that blood flow could promote the expansion of pericytes in the brain. ...
... The morpholino designed for targeting gata1a, 38 and the standard morpholino for control, were purchased from Gene Tools (Philomath, OR). Then 1 pmol nuclease-free water dissolved MO was injected into the one-cell stage embryos. ...
... To investigate the hypothesis that cardiac tissue stresses are important stimuli for trabeculation formation, we study gata1 morphant zebrafish embryos, which feature disrupted trabecular development (Galloway et al., 2005). In this model, injected morpholino oligos are used to inhibit the formation of red blood cells, resulting in a blood composition of almost entirely plasma. ...
Article
Full-text available
Cardiac trabeculae are uneven ventricular muscular structures that develop during early embryonic heart development at the outer curvature of the ventricle. Their biomechanical function is not completely understood, and while their formation is known to be mechanosensitive, it is unclear whether ventricular tissue internal stresses play an important role in their formation. Here, we performed imaging and image‐based cardiac biomechanics simulations on zebrafish embryonic ventricles to investigate these issues. Microscopy‐based ventricular strain measurements show that the appearance of trabeculae coincided with enhanced deformability of the ventricular wall. Image‐based biomechanical simulations reveal that the presence of trabeculae reduces ventricular tissue internal stresses, likely acting as structural support in response to the geometry of the ventricle. Passive ventricular pressure‐loading experiments further reveal that the formation of trabeculae is associated with a spatial homogenization of ventricular tissue stiffnesses in healthy hearts, but gata1 morphants with a disrupted trabeculation process retain a spatial stiffness heterogeneity. Our findings thus suggest that modulating ventricular wall deformability, stresses, and stiffness are among the biomechanical functions of trabeculae. Further, experiments with gata1 morphants reveal that a reduction in fluid pressures and consequently ventricular tissue internal stresses can disrupt trabeculation, but a subsequent restoration of ventricular tissue internal stresses via vasopressin rescues trabeculation, demonstrating that tissue stresses are important to trabeculae formation. Overall, we find that tissue biomechanics is important to the formation and function of embryonic heart trabeculation. image Key points Trabeculations are fascinating and important cardiac structures and their abnormalities are linked to embryonic demise. However, their function in the heart and their mechanobiological formation processes are not completely understood. Our imaging and modelling show that tissue biomechanics is the key here. We find that trabeculations enhance cardiac wall deformability, reduce fluid pressure stresses, homogenize wall stiffness, and have alignments that are optimal for providing load‐bearing structural support for the heart. We further discover that high ventricular tissue internal stresses consequent to high fluid pressures are needed for trabeculation formation through a rescue experiment, demonstrating that myocardial tissue stresses are as important as fluid flow wall shear stresses for trabeculation formation.
... The severe impairment of definitive hematopoiesis is also evident in expression analyses using RT-PCR; the results confirm the transient up-regulation of c-MYB and downregulation of GATA-1 in zebrafish (Soza-Ried et al., 2010). Erythroid precursors are converted into myeloid precursors in the loss of GATA-1 in zebrafish embryos (Galloway et al., 2005). ...
... In contrast, the myeloid progenitors (lcp1+) (ref. 31) and mature cells such as Sudan black+ (ref. 32) neutrophils were unchanged in miR-128 Δ/Δ compared with WT ( Fig. 1g and Extended Data Fig. 1b,l). ...
Article
Full-text available
Definitive haematopoietic stem and progenitor cells (HSPCs) generate erythroid, lymphoid and myeloid lineages. HSPCs are produced in the embryo via transdifferentiation of haemogenic endothelial cells in the aorta–gonad–mesonephros (AGM). HSPCs in the AGM are heterogeneous in differentiation and proliferative output, but how these intrinsic differences are acquired remains unanswered. Here we discovered that loss of microRNA (miR)-128 in zebrafish leads to an expansion of HSPCs in the AGM with different cell cycle states and a skew towards erythroid and lymphoid progenitors. Manipulating miR-128 in differentiating haemogenic endothelial cells, before their transition to HSPCs, recapitulated the lineage skewing in both zebrafish and human pluripotent stem cells. miR-128 promotes Wnt and Notch signalling in the AGM via post-transcriptional repression of the Wnt inhibitor csnk1a1 and the Notch ligand jag1b. De-repression of cskn1a1 resulted in replicative and erythroid-biased HSPCs, whereas de-repression of jag1b resulted in G2/M and lymphoid-biased HSPCs with long-term consequence on the respective blood lineages. We propose that HSPC heterogeneity arises in the AGM endothelium and is programmed in part by Wnt and Notch signalling.
... Research with the zebrafish has provided many fundamental insights into the genetic mechanisms of primitive and definitive hematopoiesis [203], from events involving hematopoietic stem cell (HSC) patterning [204] to fate choice [205,206] and differentiation [207][208][209][210]. A number of studies have contributed new insights into the effects of RA signaling on both the primitive and definitive hematopoietic waves, which are distinct in the cells they produce; namely, red blood cells and macrophages during the initial wave, followed by the production of all lineages in the subsequent wave, respectively [203]. ...
Article
Full-text available
Retinoic acid (RA) is a metabolite of vitamin A (retinol) that plays various roles in development to influence differentiation, patterning, and organogenesis. RA also serves as a crucial homeostatic regulator in adult tissues. The role of RA and its associated pathways are well conserved from zebrafish to humans in both development and disease. This makes the zebrafish a natural model for further interrogation into the functions of RA and RA-associated maladies for the sake of basic research, as well as human health. In this review, we explore both foundational and recent studies using zebrafish as a translational model for investigating RA from the molecular to the organismal scale.
Chapter
Hematopoiesis is a complex process that tightly regulates the generation, proliferation, differentiation, and maintenance of hematopoietic cells. Disruptions in hematopoiesis can lead to various diseases affecting both hematopoietic and non-hematopoietic systems, such as leukemia, anemia, thrombocytopenia, rheumatoid arthritis, and chronic granuloma. The zebrafish serves as a powerful vertebrate model for studying hematopoiesis, offering valuable insights into both hematopoietic regulation and hematopoietic diseases. In this chapter, we present a comprehensive overview of zebrafish hematopoiesis, highlighting its distinctive characteristics in hematopoietic processes. We discuss the ontogeny and modulation of both primitive and definitive hematopoiesis, as well as the microenvironment that supports hematopoietic stem/progenitor cells. Additionally, we explore the utility of zebrafish as a disease model and its potential in drug discovery, which not only advances our understanding of the regulatory mechanisms underlying hematopoiesis but also facilitates the exploration of novel therapeutic strategies for hematopoietic diseases.
Article
Tralomethrin, a synthetic pyrethroid insecticide used to control a wide range of pests in agriculture and public health, is highly toxic to aquatic organisms. However, data regarding the toxicity and underlying mechanisms of tralomethrin in aquatic organisms are limited. Thus, this study aimed to investigate the toxicity of tralomethrin in zebrafish. Zebrafish embryos were exposed to tralomethrin at different concentrations (16.63, 33.25, and 49.88 μg/L). Results showed that tralomethrin exposure caused cardiovascular dysplasia and dysfunction, including developmental abnormalities (pericardial edema, delayed yolk absorption, and uninflated swim bladder), elevated heart rate, and erythrogenesis disorders. Moreover, the expression patterns of crucial genes responsible for cardiovascular development (alas2, gata1a, hbbe2, nkx2.5, myl7, and myh6) also exhibited dysregulation in response to tralomethrin exposure. Oxidative stress occurred in embryos after exposure to tralomethrin. Collectively, our data suggest that exposure to tralomethrin induces cardiovascular and developmental toxicity in zebrafish. These findings are instrumental for evaluations of the environmental risk of tralomethrin in aquatic ecosystems in the future.
Article
Triazole fungicides (TFs) are known to be common environmental contaminants that can be toxic to aquatic animals, but their developmental toxicity is not fully understood. To address this gap, we first used a glucocorticoid receptor α (GRα)-mediated dual luciferase reporter gene system to explore the possible development toxicity of 10 TFs and found that flusilazole (FLU) exhibited stronger agonistic activity against GRα. Subsequent transcriptome sequencing showed that FLU exposure affected GRα activation and hematopoiesis associated with a variety of biological processes, including responses to corticosteroid release, embryonic hematopoiesis, erythroid differentiation, and the development of hematopoietic or lymphoid organs. Furthermore, based on in situ hybridization and staining techniques, we clarified that FLU decreased the expression of the primitive hematopoietic marker genes gata1 and pu.1. and caused the defects in the posterior blood island (PBI), thereby impacting intermediate hematopoietic processes. Also, FLU significantly reduced the expression of the crucial hematopoietic gene cmyb and disrupted the production of erythrocytes and bone marrow cells during definitive hematopoiesis. Consistently, we found that FLU induced lesions in the kidney, a hematopoietic organ, including the infiltration of inflammatory cells, tubular collapse, reduced tubular filtration area, and interstitial hydronephrosis. We also found that FLU increased aberrant red blood cells in the peripheral blood of zebrafish. These findings provide new insights into the developmental toxicity and ecotoxicological risk of TFs.
Article
Full-text available
In vertebrates, hematopoietic and vascular progenitors develop from ventral mesoderm. The first primitive wave of hematopoiesis yields embryonic red blood cells, whereas progenitor cells of subsequent definitive waves form all hematopoietic cell lineages. In this report we examine the development of hematopoietic and vasculogenic cells in normal zebrafish and characterize defects inclocheandspadetailmutant embryos. The zebrafish homologs oflmo2, c-myb, fli1, flk1, andflt4have been cloned and characterized in this study. Expression of these genes identifies embryonic regions that contain hematopoietic and vascular progenitor cells. The expression ofc-mybalso identifies definitive hematopoietic cells in the ventral wall of the dorsal aorta. Analysis ofb316mutant embryos that carry a deletion of thec-mybgene demonstrates thatc-mybis not required for primitive erythropoiesis in zebrafish even though it is expressed in these cells. Bothclocheandspadetailmutant embryos have defects in primitive hematopoiesis and definitive hematopoiesis. Theclochemutants also have significant decreases in vascular gene expression, whereasspadetailmutants expressed normal levels of these genes. These studies demonstrate that the molecular mechanisms that regulate hematopoiesis and vasculogenesis have been conserved throughout vertebrate evolution and thecloandsptgenes are key regulators of these programs.
Article
Full-text available
GATA-1 is a zinc-finger transcription factor believed to play an important role in gene regulation during the development of erythroid cells, megakaryocytes and mast cells. Other members of the GATA family, which can bind to the same DNA sequence motif, are co-expressed in several of these hemopoietic lineages, raising the possibility of overlap in function. To examine the specific roles of GATA-1 in hematopoietic cell differentiation, we have tested the ability of embryonic stem cells, carrying a targeted mutation in the X-linked GATA-1 gene, to contribute to various blood cell types when used to produce chimeric embryos or mice. Previously, we reported that GATA-1- mutant cells failed to contribute to the mature red blood cell population, indicating a requirement for this factor at some point in the erythroid lineage (L. Pevny et al., (1991) Nature 349, 257-260). In this study, we have used in vitro colony assays to identify the stage at which mutant erythroid cells are affected, and to examine the requirement for GATA-1 in other lineages. We found that the development of erythroid progenitors in embryonic yolk sacs was unaffected by the mutation, but that the cells failed to mature beyond the proerythroblast stage, an early point in terminal differentiation. GATA-1- colonies contained phenotypically normal macrophages, neutrophils and megakaryocytes, indicating that GATA-1 is not required for the in vitro differentiation of cells in these lineages. GATA-1- megakaryocytes were abnormally abundant in chimeric fetal livers, suggesting an alteration in the kinetics of their formation or turnover. The lack of a block in terminal megakaryocyte differentiation was shown by the in vivo production of platelets expressing the ES cell-derived GPI-1C isozyme. The role of GATA-1 in mast cell differentiation was examined by the isolation of clonal mast cell cultures from chimeric fetal livers. Mutant and wild-type mast cells displayed similar growth and histochemical staining properties after culture under conditions that promote the differentiation of cells resembling mucosal or serosal mast cells. Thus, the mast and megakaryocyte lineages, in which GATA-1 and GATA-2 are co-expressed, can complete their maturation in the absence of GATA-1, while erythroid cells, in which GATA-1 is the predominant GATA factor, are blocked at a relatively early stage of maturation.
Article
GATA-1 germline mutation in mice results in embryonic lethality due to defective erythroid cell maturation, and thus other hematopoietic GATA factors do not compensate for the loss of GATA-1. To determine whether the obligate presence of GATA-1 in erythroid cells is due to its distinct biochemical properties or spatiotemporal patterning, we attempted to rescue GATA-1 mutant mice with hematopoietic GATA factor complementary DNAs (cDNAs) placed under the transcriptional control of the GATA-1gene. We found that transgenic expression of a GATA-1 cDNA fully abrogated the GATA-1–deficient phenotype. Surprisingly, GATA-2 and GATA-3 factors expressed from the same regulatory cassette also rescued the embryonic lethal phenotype of the GATA-1 mutation. However, adult mice rescued with the latter transgenes developed anemia, while GATA-1 transgenic mice did not. These results demonstrate that the transcriptional control dictating proper GATA-1 accumulation is the most critical determinant of GATA-1 activity during erythropoiesis. The results also show that there are biochemical distinctions among the hematopoietic GATA proteins and that during adult hematopoiesis the hematopoietic GATA factors are not functionally equivalent.
Article
The zinc-finger transcription factor GATA-2 plays a critical role in maintaining the pool of early hematopoietic cells. To define its specific functions in the proliferation, survival, and differentiation of hematopoietic cells, we analyzed the hematopoietic potential of GATA-2−/− cells in in vitro culture systems for proliferation and maintenance of uncommitted progenitors or differentiation of specific lineages. From a two-step in vitro differentiation assay of embryonic stem cells and in vitro culture of yolk sac cells, we demonstrate that GATA-2 is required for the expansion of multipotential hematopoietic progenitors and the formation of mast cells, but dispensable for the terminal differentiation of erythroid cells and macrophages. The rare GATA-2−/− multipotential progenitors that survive proliferate poorly and generate small colonies with extensive cell death, implying that GATA-2 may play a role in both the proliferation and survival of early hematopoietic cells. To explore possible mechanisms resulting in the hematopoietic defects of GATA-2−/− cells, we interbred mutant mouse strains to assess the effects of p53 loss on the behavior of GATA-2−/− hematopoietic cells. Analysis of GATA-2−/−/p53−/− compound-mutant embryos shows that the absence of p53 partially restores the number of total GATA-2−/− hematopoietic cells, and therefore suggests a potential link between GATA-2 and p53 pathways.
Article
Mouse embryonic stem (ES) cells lacking the transcription factor GATA-1 do not produce mature red blood cells either in vivo or in vitro. To define the consequences of GATA-1 loss more precisely, we used an in vitro ES cell differentiation assay that permits enumeration of primitive (EryP) and definitive (EryD) erythroid precursors and recovery of pure erythroid colonies. In contrast to normal ES cells, GATA-1- ES cells fail to generate EryP precursors. EryD precursors, however, are normal in number but undergo developmental arrest and death at the proerythroblast stage. Contrary to initial expectations, arrested GATA-1(-)-definitive proerythroblasts express GATA target genes at normal levels. Transcripts of the related factor GATA-2 are remarkably elevated in GATA-1- proerythroblasts. These findings imply substantial interchangeability of GATA factors in vivo and suggest that GATA-1 normally serves to repress GATA-2 during erythropoiesis. The approach used here is a paradigm for the phenotypic analysis of targeted mutations affecting hematopoietic development.
Article
The X chromosome-linked transcription factor GATA-1 is expressed specifically in erythroid, mast, megakaryocyte, and eosinophil lineages, as well as in hematopoietic progenitors. Prior studies revealed that gene-disrupted GATA-1- embryonic stem cells give rise to adult (or definitive) erythroid precursors arrested at the proerythroblast stage in vitro and fail to contribute to adult red blood cells in chimeric mice but did not clarify a role in embryonic (or yolk sac derived) erythroid cells. To examine the consequences of GATA-1 loss on embryonic erythropoiesis in vivo, we inactivated the GATA-1 locus in embryonic stem cells by gene targeting and transmitted the mutated allele through the mouse germ line. Male GATA-1- embryos die between embryonic day 10.5 and 11.5 (E10.5-E11.5) of gestation. At E9.5, GATA-1- embryos exhibit extreme pallor yet contain embryonic erythroid cells arrested at an early proerythroblast-like stage of their development. Embryos stain weakly with benzidine reagent, and yolk sac cells express globin RNAs, indicating globin gene activation in the absence of GATA-1. Female heterozygotes (GATA-1+/-) are born pale due to random inactivation of the X chromosome bearing the normal allele. However, these mice recover during the neonatal period, presumably as a result of in vivo selection for progenitors able to express GATA-1. Our findings conclusively establish the essential role for GATA-1 in erythropoiesis within the context of the intact developing mouse and further demonstrate that the block to cellular maturation is similar in GATA-1- embryonic and definitive erythroid precursors. Moreover, the recovery of GATA-1+/- mice from anemia seen at birth provides evidence indicating a role for GATA-1 at the hematopoietic progenitor cell level.