• Zebrafish flt3 plays an important role in the initiation of definitive hematopoietic stem cells.

  • Expression of human FLT3-ITD activates endogenous flt3 signaling and induces myeloid expansion.

FMS-like tyrosine kinase 3 (FLT3) is expressed in human hematopoietic stem and progenitor cells (HSPCs) but its role during embryogenesis is unclear. In acute myeloid leukemia (AML), internal tandem duplication (ITD) of FLT3 at the juxtamembrane (JMD) and tyrosine kinase (TKD) domains (FLT3-ITD+) occurs in 30% of patients and is associated with inferior clinical prognosis. TKD mutations (FLT3-TKD+) occur in 5% of cases. We made use of zebrafish to examine the role of flt3 in developmental hematopoiesis and model human FLT3-ITD+ and FLT3-TKD+ AML. Zebrafish flt3 JMD and TKD were remarkably similar to their mammalian orthologs. Morpholino knockdown significantly reduced the expression of l-plastin (pan-leukocyte), csf1r, and mpeg1 (macrophage) as well as that of c-myb (definitive HSPCs), lck, and rag1 (T-lymphocyte). Expressing human FLT3-ITD in zebrafish embryos resulted in expansion and clustering of myeloid cells (pu.1+, mpo+, and cebpα+) which were ameliorated by AC220 and associated with stat5, erk1/2, and akt phosphorylation. Human FLT3-TKD (D835Y) induced significant, albeit modest, myeloid expansion resistant to AC220. This study provides novel insight into the role of flt3 during hematopoiesis and establishes a zebrafish model of FLT3-ITD+ and FLT3-TKD+ AML that may facilitate high-throughput screening of novel and personalized agents.

The zebrafish has emerged as a model for the study of embryonic hematopoiesis.1,2  In primitive hematopoiesis, pu.1 is first expressed at 12 hours postfertilization (hpf) in the anterior lateral plate mesoderm (ALPM) where it plays a role in driving cellular differentiation toward myeloid fate, independent of primitive erythropoiesis that occurs in the posterior lateral plate mesoderm.3  After formation of the rostral blood island at 16 hpf, the pu.1+ myeloid progenitor cells begin to spread on the yolk sac,4  and subsequently switch on expression of pan-leukocyte gene l-plastin,5-7  as well as those genes associated with macrophage (csf1ra, mfap4, cxcr3.2, mpeg1, and ptpn6)8  and neutrophil lineages (lyc and mpo).5  The erythromyeloid progenitors that arise autonomously in the posterior blood island (PBI) also generate myeloid cells associated with l-plastin, pu.1, and mpo expression at 30 hpf.9 

Definitive hematopoiesis arises from the ventral wall of the dorsal aorta (DA) as evidenced by the expression of c-myb and runx-1 at 36 hpf.9,10  It subsequently migrates to the caudal hematopoietic tissue (CHT), thence the kidney marrow, where lifelong hematopoiesis occurs.11 gcsf and cebpα are important regulators of granulopoiesis.12 

Recent studies also demonstrated that myeloid and erythroid cell fates from the myeloerythroid progenitor cells are precisely controlled by the interplay between pu.1 and gata112,13  and the sumoylation of cebpα promotes myeloid cell expansion at the expense of erythroid development.14 

FMS-like tyrosine kinase 3 (FLT3) is a class III receptor tyrosine kinase normally expressed in human hematopoietic stem and progenitor cells (HSPCs).15  FLT3 consists of 5 immunoglobulin-like motifs in the extracellular domain, a transmembrane domain, a juxtamembrane domain (JMD), and 2 tyrosine kinase domains (TKDs) in the intracellular region.16,17  Under normal circumstances, binding of FLT3 to its ligand results in FLT3 homodimerization and phosphorylation of TKD and hence activation of multiple effectors, providing the proliferative and survival signals to HSPCs.16-19  Flt3 function on hematopoiesis has been evaluated by targeted deletion in mice and serial transplantations. Specifically, Flt3 knockout was compatible with postnatal survival with only defective B-lymphoid development at baseline.20  In competitive and serial transplantation, although T-lymphoid and myeloid reconstitutions were reduced in primary recipients, they remained stable in secondary recipients. A more recent study only revealed defective B-lymphoid but no effect on hematopoietic stem cell (HSC), T-lymphoid, or myeloid reconstitution upon primary and secondary transplantations.21  These observations supported the proposition that Flt3 signaling is dispensable for HSC self-renewal. The role of Flt3 during embryonic hematopoiesis is currently unclear.

Internal tandem duplication (ITD) at the JMD and TKD of FLT3 occurs in 30% of human acute myeloid leukemia (AML) and is associated with inferior clinical prognosis. Single-base mutations at TKD occur in 5% of cases whose prognostic significance is less clear.22  These gain-of-function changes resulted in constitutive activation of FLT3 and downstream effectors including signal transducer and activator of transcription 5 (STAT5),23  AKT,24  and extracellular signal-regulated kinase (ERK).25  Newer tyrosine kinase inhibitors (TKIs) including sorafenib and AC220 were effective in inducing remission. However, the effects were transient and emergence of mutant clones, particularly at the residue 835 of the TKD, where aspartate was replaced by tyrosine (D835Y) hence conferring resistance to TKI, was common,26,27  suggesting that the targets are likely to be evolving28  and that a robust platform and animal model to identify more effective and personalized TKI in real time is needed.

We made use of the zebrafish model to examine the role of flt3 in primitive and definitive hematopoiesis during embryonic development and validate a novel zebrafish model overexpressing human FLT3-ITD and FLT3-TKD with a view of developing a whole-organism platform for personalizing TKI treatment of AML.

Zebrafish maintenance and embryo collection

Wild-type (WT) zebrafish were maintained under standard conditions and embryos were staged as described.29,30  The study was approved by the Committee of the Use of Live Animals for Teaching and Research in The University of Hong Kong.

Cloning of zebrafish flt3 gene

Total RNA was extracted (TRIzol; Invitrogen) from AB zebrafish embryos at 1 day postfertilization (dpf), and reverse transcribed into complementary DNA (cDNA) (SuperScript II Reverse Transcriptase; Invitrogen). A pair of primers (supplemental Table 1, see supplemental Data available on the Blood Web site) was designed to amplify the coding sequence of flt3 sequence by polymerase chain reaction (PCR) using Ex-Taq DNA polymerase (TaKaRa Biotechnology), cloned into pBluescript II SK(+) vector by EcoRI and BamHI and bidirectionally sequenced (Techdragon Limited, for all sequencing in this study) by primer walking (supplemental Table 1).

MO-mediated flt3 gene knockdown

The morpholino (MO; Gene Tools, LLC) technology has been described.31,32 flt3 MO (6 ng) targeting the 5′ untranslated regions (UTRs) (supplemental Table 1) was microinjected into 1-cell–stage embryos. The zebrafish flt3 UTR (containing the MO targeting sequence) and partial coding sequence (supplemental Table 1) was cloned into the pegfp-N3 vector (Clontech/BD Biosciences) to generate a flt3UTR-egfp fusion chimeric gene.

WISH assay

The partial cDNA (642 bp) sequence of flt3 was amplified by PCR (supplemental Table 1) and the PCR products were cloned into a pGEM-T-easy vector (Promega) and used as template to generate an antisense digoxigenin (DIG)–labeled RNA probe (DIG RNA Labeling Kit; Roche Applied Science) in vitro.33,34  Double whole-mount in situ hybridization (WISH) was performed using the DIG- and fluorescein isothiocyanate–labeled probe as previously described.35 

Real-time quantitative PCR analysis

Total RNA from 30 embryos was isolated using TRIzol reagent (Life Technologies) and reverse transcribed (SuperScript II Reverse Transcriptase; Invitrogen). Primers were shown in supplemental Table 1. Reverse transcription PCR has been described previously.36,37 

Whole-mount phospho-Histone H3 (Ser10) immunostaining

Immunostaining was performed using anti-phospho-Histone H3 (Upstate) with slight modifications38  (for details, refer to the supplemental Figure 7 legend).

Synthesis of zebrafish flt3 messenger RNA for rescue experiments

Total RNA of AB zebrafish embryos at 24 hpf was extracted and reversed transcribed. Two primers (supplemental Table 1) were used to amplify the flt3 open reading frame. The PCR product was cloned into the pBluescript II SK(+) vector (pBSK-flt3) and confirmed by bidirectional sequencing. The pBSK-flt3 vector was linearized by SacII, purified (QIAquick PCR Purification Kit; QIAGEN), and used for in vitro transcription by T7 RNA polymerase (mMESSAGE mMACHINE T7 Kit; Ambion).

Overexpression of human FLT3 mutation in zebrafish

Total RNA from AML cell line KG-1 carrying WT FLT3 was extracted and reverse transcribed. pMSCV-neo-FLT3-ITD containing human FLT-ITD (21 bp duplication) was subcloned. WT FLT3 and FLT3-ITD were amplified using a reverse primer containing the T2a self-cleaving peptide sequence (supplemental Table 1) and inserted into the pegfp-N3 vector in frame with the egfp gene. The plasmids pegfp-N3, pHsFLT3-WT-T2a-egfp, and pHsFLT3-ITD-T2a-egfp were linearized by StuI and purified. Human FLT3-ITD was in-frame cloned into the pDsRed-Monomer-N1 vector to generate the FLT3-ITD-T2a-mRFP fusion gene (Clontech/BD Sciences). Human FLT3-ITD was also cloned into pBluescript II SK(+), and the human FLT3-ITD messenger RNA (mRNA) was in vitro transcribed for microinjection (300 pg per embryo).

Generation of FLT3-TKD mutation by site-directed mutagenesis assay

Site-directed mutagenesis was performed to generate the FLT3-TKD and FLT3-ITD-TKD (D835Y) mutation (supplemental Table 1). Plasmid (100 ng) and specific primers (1.25 pmol each) were used in PCR with 18 cycles using Platinum Pfx DNA polymerase platform. Cloning and Sanger sequencing followed standard protocols.

Pharmacologic inhibition of FLT3 in zebrafish embryos

Sorafenib and sunitinib (LC Laboratories) were multikinase inhibitors used clinically for the treatment of hepatocellular and renal cell carcinomas.39,40  AC220 (LC Laboratories) is a potent and relatively selective inhibitor of FLT3 currently evaluated in clinical trials for AML treatment.41  Embryos after gastrulation (6 hpf) were treated with drugs until collection for examination. Equivolume amounts of dimethylsulfoxide (DMSO) were used in parallel experiments using the same clutch of embryos as control.

Microscopy and imaging

The fluorescent images of embryos were taken under Olympus IX70 (Olympus Corporation) and a 10×/0.3 NA objective in 3% methylcellulose, with Olympus DP71 (Olympus Corporation) and Olympus DP-BSW basic software, processed with Adobe Photoshop (version 8.0.1) Bright-field images of embryos were taken under Nikon SMZ800 (Nikon Hong Kong Ltd.) and a P-Plan 1× objective in 3% methylcellulose with Nikon Digital Sight DS-Fil (Nikon Hong Kong Ltd.) and processed with Adobe Photoshop (version 8.0.1) and Image J (National Institutes of Health).

Statistical analysis

Data were expressed as means ± standard error of the mean. Comparisons between groups of numerical data were evaluated using the Student t test. Categorical data were evaluated using the χ2 test. P values < .05 were considered statistically significant, and were represented with an asterisk (*P < .05 or **P < .01).

FLT3 is highly conserved between zebrafish and mammal

Zebrafish flt3 contained 2871 bp and encoded a 956-aa protein (supplemental Figure 1). FLT3 are found in different species (supplemental Figure 2) and full-length flt3 protein shared an overall 32%, 35%, and 34% sequence identity with those of human, mouse, and rat, respectively. The JMD and the activation loops of TKD are highly conserved (Figure 1A), implicating conserved functions of FLT3 signaling during evolution.

Figure 1

Conservation, expression, and knockdown of zebrafish flt3. (A) Multiple sequence alignment showing the conservation of JMD and the activation loop within the TKD in FLT3 from zebrafish (Dr, Danio rerio), human (Hs, Homo sapiens), mouse (Mm, Mus musculus), and rat (Rn, Rattus norvegicus). (B-E) WISH showing the spatial expression of flt3 at 3 (B), 5 (C), 12 (D), and 18 (E) hpf. (F-G) Double ISH (flt3 in blue; α-eHb1 in red) showing flt3 expression in the homozygous cloche mutant (F) and WT and heterozygous siblings (G) at 18 hpf. Inset, A higher magnification of region in the anterior yolk sac as defined by the rectangle. (H-K) WISH (H-I) and paraffin sectioning (J-K) showing flt3 expression at 28 hpf. Sectioning on the flt3 sense probe ISH embryos (inset) was used as negative control. (L) ISH showing flt3 expression in the PBI at 32 hpf. (M-O) The egfp expression in flt3UTR-egfp injected (M) and flt3UTR-egfp + flt3MO coinjected (N) embryos at 2 dpf, respectively. Comparison of the percentage (O) of embryos showing the egfp expression with or without the coinjection of flt3MO. (P-Q) General morphology of uninjected control (P) and flt3MO (Q) embryos at 2 dpf. Triangle in panel A indicates the human FLT3 D835 residue; the arrows in panel I indicate the flt3 expression in the midline of the embryos; the arrows in panel L indicated the flt3 expression in the PBI. Scale bars represent 500 μm. ISH, in situ hybridization; NC, notochord; PCV, posterior cardinal vein.

Figure 1

Conservation, expression, and knockdown of zebrafish flt3. (A) Multiple sequence alignment showing the conservation of JMD and the activation loop within the TKD in FLT3 from zebrafish (Dr, Danio rerio), human (Hs, Homo sapiens), mouse (Mm, Mus musculus), and rat (Rn, Rattus norvegicus). (B-E) WISH showing the spatial expression of flt3 at 3 (B), 5 (C), 12 (D), and 18 (E) hpf. (F-G) Double ISH (flt3 in blue; α-eHb1 in red) showing flt3 expression in the homozygous cloche mutant (F) and WT and heterozygous siblings (G) at 18 hpf. Inset, A higher magnification of region in the anterior yolk sac as defined by the rectangle. (H-K) WISH (H-I) and paraffin sectioning (J-K) showing flt3 expression at 28 hpf. Sectioning on the flt3 sense probe ISH embryos (inset) was used as negative control. (L) ISH showing flt3 expression in the PBI at 32 hpf. (M-O) The egfp expression in flt3UTR-egfp injected (M) and flt3UTR-egfp + flt3MO coinjected (N) embryos at 2 dpf, respectively. Comparison of the percentage (O) of embryos showing the egfp expression with or without the coinjection of flt3MO. (P-Q) General morphology of uninjected control (P) and flt3MO (Q) embryos at 2 dpf. Triangle in panel A indicates the human FLT3 D835 residue; the arrows in panel I indicate the flt3 expression in the midline of the embryos; the arrows in panel L indicated the flt3 expression in the PBI. Scale bars represent 500 μm. ISH, in situ hybridization; NC, notochord; PCV, posterior cardinal vein.

Close modal

Gene expression profiling of zebrafish flt3

In adult fish, flt3 expression was found in gill, heart, spleen, and kidney (supplemental Figure 3A). At the embryonic stage, flt3 mRNA was detected as early as the 1-cell stage, indicating maternal transcripts (supplemental Figure 3B). Subsequently, flt3 expression increased from 6 hpf, peaked at 24 hpf, and maintained up to 48 hpf. WISH demonstrated ubiquitous flt3 expression at 3, 5, and 12 hpf (Figure 1B-D). The ubiquitous expression of flt3 was confirmed by histologic sectioning of embryos at 12 hpf (supplemental Figure 4A-B). At 18 hpf, expression at the anterior yolk sac could be demonstrated (Figure 1E). To further ascertain the hematopoietic origin of flt3-expressing cells, the cloche mutants in which primitive hematopoiesis was defective were examined. Normally, α-eHb1 was robustly expressed at the intermediate cell mass (ICM) of WT and heterozygous cloche embryos, and its absence served to ascertain the homozygous state of the cloche embryos. Based on this differential α-eHb1 expression, we demonstrated that flt3 was expressed in the WT and heterozygous cloche embryos (51 of 70; 72.86%) but not in the homozygous mutants (19 of 70; 27.14%) (Figure 1F-G; supplemental Figure 4C-J). At 24 hpf, flt3 was predominantly expressed in the cardiac mesoderm and pronephric ducts (supplemental Figure 4K). At 28 hpf, flt3 was expressed, albeit weakly, in the embryonic midline that included the region of the ventral wall of DA as shown in both WISH and paraffin sections (Figure 1H-K). At 32 hpf, flt3 was expressed in a small cluster of cells in the PBI (Figure 1L). At 48 hpf, flt3 expression was found near the developing heart and gut, which became more prominent at 72 hpf. On 4 dpf, its expression in the heart region disappeared and became restricted in the pharynx and intestine regions (supplemental Figure 4L-N).

Knockdown of flt3 perturbed primitive myelopoiesis

Embryos were injected with MO (flt3MO) targeting the 5′-UTR of the flt3 gene. The embryos could tolerate up to 6 ng without severe toxicity and this dose was used throughout the study. To ascertain the molecular targeting, we injected a flt3UTR-egfp chimeric gene containing the flt3MO targeting site in the 5′ terminus (supplemental Figure 5) and demonstrated mosaic egfp expression (Figure 1M). The latter could be quenched by flt3MO (Figure 1N-O; flt3UTR-egfp, 53.03% ± 7.93%; flt3UTR-egfp + MO, 5.10% ± 0.92%, P = .008). The majority of flt3 morphants had no significant developmental defects up to 2 dpf (Figure 1P-Q). Less than 10% of embryos showed deformity with curved tail, pericardial edema, and developmental delay and were excluded from subsequent analysis. Among embryos with normal morphology, l-plastin (Figure 2A-B; supplemental Figure 6M) but not pu.1 (supplemental Figure 6I-J) expression in the anterior yolk sac region was significantly downregulated at 18 hpf. At 22 hpf, both csf1r and mpeg1 expression, which denote macrophage, were significantly reduced (supplemental Figure 6O-P). mpo expression was only modestly reduced (supplemental Figure 6N). These observations suggested that flt3 knockdown more specifically perturbed macrophage derivation during primitive myelopoiesis. Importantly, a proportion of primitive macrophages in the anterior yolk sac in the control embryos (injected with scramble sequence MO) underwent proliferation as shown by overlapping signals of egfp (indicative of mpeg1 expression in macrophages) and Alexa Fluor 594 (indicative of cellular proliferation with positive phospho-histone H3 [Ser10] immunostaining) (Figure 2D-F and supplemental Figure 7D-F, white arrow). In contrast, the primitive macrophages in flt3 morphant showed significantly reduced proliferative signals (Figure 2C,G-I; supplemental Figure 7H-J). Proliferative cells (nonmacrophage) were also identified in the heads and tail regions (supplemental Figure 7G,K). There was no change in apoptosis as shown by TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling) staining in these primitive macrophages (supplemental Figure 7L-S). Therefore, these observations provided unequivocal evidence that flt3 knockdown inhibited proliferation of primitive macrophages during embryonic development. The effects were specific to myeloid development but not a general one on ALPM as scl and lmo2 expression in ALPM were unaffected (supplemental Figure 6A-D). These observations supported the proposition that flt3 signaling is important for the maintenance of primitive myelopoiesis after their myeloid commitment driven by pu.1. Genes associated with erythropoiesis (gata1, α-eHb1) in ICM were also unaffected (supplemental Figure 6E-H). Therefore, an effect of flt3 on cellular specification and myeloid-erythroid skewing was less likely.

Figure 2

flt3 knockdown affected primitive myelopoiesis and definitive hematopoiesis. MO targeting zebrafish flt3 was microinjected into 1-cell–stage embryos (6 ng per embryo), and uninjected embryos from the same batch were used as control. (A) WISH comparing the expression of l-plastin between control (top panel) and flt3MO embryos (bottom panel) at 18 hpf. The arrow indicated l-plastin expression at the anterior yolk sac region. (B) Comparison of l-plastin expression (dots in the anterior yolk sac) in control and flt3MO embryos at 18 hpf (corresponding to panel A). (C) Comparison of percentage of Phospho-Histone H3 (pH3) positive macrophages in control and flt3MO embryos. The percentage was calculated based on the number of overlapped signals divided by the number of macrophages (GFP+) in each embryo. The results represented mean ± 1 standard error of the mean of 15 embryos examined in 3 different experiments. (D-I) Phospho-Histone H3 (Ser10) immunostaining showing the proliferation of macrophages (mpeg1:egfp transgenic fish) in control embryos (D-F) and flt3 morphants (G-I) at 22 hpf. The images in panels D-F were obtained from the same embryo, showing the overlapping signals in panel F. It was similarly presented for panels G-I. The body including the developing eyes and yolk sac was outlined by the dotted lines. Proliferative cells (nonmacrophage) were also identified in the heads and tail regions and were shown in supplemental Figure 7G,K. (J-P) WISH showing the c-myb expression in control (J,L,O) and flt3MO (K,M,P) embryos at 36, 72, and 96 hpf, respectively. The arrows in panel J and arrowheads in panels L and O indicated c-myb expression in the ventral wall of DA and CHT. (N) Real-time quantitative PCR comparing c-myb expression between control and flt3MO embryos (corresponding to panels L-M). (Q-S) Comparison of cd41:egfp expression in CHT between control (Q) and flt3MO (R) embryos using Tg(cd41:egfp) transgenic embryos. (S) Comparison of cd41:egfp expression (dots of egfp+ cells in the CHT) in control and flt3MO embryos (corresponding to panels Q-R). (T-U) WISH showing rag1 expression in control (T) and flt3MO (U) embryos. The circle in panel T and arrow in the inset indicated rag1 expression in the thymus at lateral and ventral views. (V-W) Comparison of lck expression in thymus between control (V) and flt3MO (W) embryos using Tg(lck:egfp) transgenic embryos. The circle in panel V indicated lck expression in the thymus. Scale bars represent 500 μm.

Figure 2

flt3 knockdown affected primitive myelopoiesis and definitive hematopoiesis. MO targeting zebrafish flt3 was microinjected into 1-cell–stage embryos (6 ng per embryo), and uninjected embryos from the same batch were used as control. (A) WISH comparing the expression of l-plastin between control (top panel) and flt3MO embryos (bottom panel) at 18 hpf. The arrow indicated l-plastin expression at the anterior yolk sac region. (B) Comparison of l-plastin expression (dots in the anterior yolk sac) in control and flt3MO embryos at 18 hpf (corresponding to panel A). (C) Comparison of percentage of Phospho-Histone H3 (pH3) positive macrophages in control and flt3MO embryos. The percentage was calculated based on the number of overlapped signals divided by the number of macrophages (GFP+) in each embryo. The results represented mean ± 1 standard error of the mean of 15 embryos examined in 3 different experiments. (D-I) Phospho-Histone H3 (Ser10) immunostaining showing the proliferation of macrophages (mpeg1:egfp transgenic fish) in control embryos (D-F) and flt3 morphants (G-I) at 22 hpf. The images in panels D-F were obtained from the same embryo, showing the overlapping signals in panel F. It was similarly presented for panels G-I. The body including the developing eyes and yolk sac was outlined by the dotted lines. Proliferative cells (nonmacrophage) were also identified in the heads and tail regions and were shown in supplemental Figure 7G,K. (J-P) WISH showing the c-myb expression in control (J,L,O) and flt3MO (K,M,P) embryos at 36, 72, and 96 hpf, respectively. The arrows in panel J and arrowheads in panels L and O indicated c-myb expression in the ventral wall of DA and CHT. (N) Real-time quantitative PCR comparing c-myb expression between control and flt3MO embryos (corresponding to panels L-M). (Q-S) Comparison of cd41:egfp expression in CHT between control (Q) and flt3MO (R) embryos using Tg(cd41:egfp) transgenic embryos. (S) Comparison of cd41:egfp expression (dots of egfp+ cells in the CHT) in control and flt3MO embryos (corresponding to panels Q-R). (T-U) WISH showing rag1 expression in control (T) and flt3MO (U) embryos. The circle in panel T and arrow in the inset indicated rag1 expression in the thymus at lateral and ventral views. (V-W) Comparison of lck expression in thymus between control (V) and flt3MO (W) embryos using Tg(lck:egfp) transgenic embryos. The circle in panel V indicated lck expression in the thymus. Scale bars represent 500 μm.

Close modal

Knockdown of zebrafish flt3 reduced definitive hematopoiesis

The effect of flt3 knockdown on definitive hematopoiesis was also examined. c-myb expression was consistently downregulated in the ventral wall of DA at 36 hpf (Figure 2J-K) and the CHT at 72 (Figure 2L-N) and 96 hpf (Figure 2O-P). cd41:egfp+ cells in CHT of Tg(cd41:egfp) flt3 morphants were also significantly reduced at 72 hpf (Figure 2Q-S). Moreover, the expression of recombination activating gene 1 (Figure 2T-U, rag1) and T-cell–specific light-chain kinase (Figure 2V-W, lck) in the thymus were also downregulated. To confirm the specificity of these phenotypes, downregulation of c-myb and rag1 by flt3MO (percentage of embryos with normal gene expression in morphants: c-myb: 15.0% ± 3.6%; rag1: 13.2% ± 0.7%) could be rescued by coinjecting the embryos with WT flt3 mRNA (Figure 3A-B) (c-myb: 75.4% ± 7.4%; P = .008; rag1, 78.7% ± 3.2%; P = .002, N = 3 experiments).

Figure 3

flt3 mRNA rescue and flt3 knockdown had no effect on angiogenesis and vasculogenesis. The expression of c-myb (A) and rag1 (B) in uninjected control, flt3MO, and flt3MO + flt3 mRNA injected embryos at 4 dpf. The arrowheads in panel Ai and the arrow in Bi indicated c-myb (CHT region) and rag1 (thymus) expression. (C) The development of ISV, DA, and DV in control and flt3MO embryos at 24 hpf. (D) flt4 (DV) and efnb2α (DA) expression in control and flt3MO embryos at 32 hpf. Panel Ai-iii represents the whole mount in situ hybridization of c-myb in control (Ai), flt3MO (ii), and flt3MO rescue (iii) embryos at 96 hpf, respectively. Panel Bi-iii represents the whole mount in situ hybridization of rag1 in control (Bi), flt3MO (ii), and flt3MO rescue (iii) at 96 hpf, respectively. Panel Ci-iv represents the green fluorescence (indicates the flk1 expression) in control (Ci and iii), and flt3MO (ii and iv) embryos at 24 hpf, respectively. Panel Di-iv represents the flt4 (Di and ii) and efnb2α expression in control (Di, iii) and flt3MO (ii, iv) at 32 hpf, respectively. Scale bars represent 250 μm.

Figure 3

flt3 mRNA rescue and flt3 knockdown had no effect on angiogenesis and vasculogenesis. The expression of c-myb (A) and rag1 (B) in uninjected control, flt3MO, and flt3MO + flt3 mRNA injected embryos at 4 dpf. The arrowheads in panel Ai and the arrow in Bi indicated c-myb (CHT region) and rag1 (thymus) expression. (C) The development of ISV, DA, and DV in control and flt3MO embryos at 24 hpf. (D) flt4 (DV) and efnb2α (DA) expression in control and flt3MO embryos at 32 hpf. Panel Ai-iii represents the whole mount in situ hybridization of c-myb in control (Ai), flt3MO (ii), and flt3MO rescue (iii) embryos at 96 hpf, respectively. Panel Bi-iii represents the whole mount in situ hybridization of rag1 in control (Bi), flt3MO (ii), and flt3MO rescue (iii) at 96 hpf, respectively. Panel Ci-iv represents the green fluorescence (indicates the flk1 expression) in control (Ci and iii), and flt3MO (ii and iv) embryos at 24 hpf, respectively. Panel Di-iv represents the flt4 (Di and ii) and efnb2α expression in control (Di, iii) and flt3MO (ii, iv) at 32 hpf, respectively. Scale bars represent 250 μm.

Close modal

Zebrafish flt3 knockdown had no effect on angiogenesis and vasculogenesis

To ascertain whether the effects of flt3 knockdown on definitive HSPCs could result from perturbation of angiogenesis and vasculogenesis,42  these processes were examined in the flt3 morphants. In Tg(flk1:egfp) embryos, the intersegmental vessels (ISVs), the DA, and dorsal vein (DV) were intact at 24 hpf after flt3 knockdown (Figure 3C). The expression of flt4 (vein) and efnb2a (artery) was also unaffected (Figure 3D).

Inhibition of zebrafish flt3 recapitulated the phenotypes of flt3 morphants

AC220 is a potent and relatively selective inhibitor of FLT3 and is currently being evaluated in clinical trials for the treatment of AML.41,43  We tested whether AC220 could recapitulate the hematopoietic phenotypes of the flt3 morphants. AC220 (2.5 μmol/L) had no effect on embryo morphology but significantly reduced the expression of l-plastin in the anterior yolk sac at 18 hpf (Figure 4A-C) and c-myb in the ventral wall of DA at 32 (Figure 4D-E) and 96 hpf (Figure 4J-K) as well as rag1 in the thymus at 96 hpf (Figure 4L-M). Vasculogenesis, as shown by flt4 (Figure 4F-G, DV) and efnb2α (Figure 4H-I, DA), expression was intact.

Figure 4

flt3 inhibitor recapitulated the effect of flt3 MO knockdown. AC220 (2.5 μmol/L) was used to treat the embryos from 6 hpf to defined developmental stage. Equal volume of DMSO treatment was used as control. (A-B) WISH comparing the l-plastin expression in the anterior yolk sac region between DMSO control (A) and AC220 (B) embryos at 19 hpf. (C) Comparison of l-plastin expression in DMSO control and AC220 embryos at 19 hpf. (D-I) WISH comparing the expression of c-myb (D-E), flt4 (F-G), and efnb2α (H-I) between DMSO control (D,F,H) and AC220 (E,G,I) embryos at 32 hpf. The arrowheads in panel D indicated c-myb expression along the ventral wall of DA. (J-M) WISH showing the expression of c-myb (J-K) and rag1 (L-M) expression in DMSO control (J,L) and AC220 (K,M) embryos at 96 hpf. The arrow and arrowheads in panel J indicated c-myb expression at the thymus and CHT. The circled region (L-M) indicates the rag1 expression in the thymus. Scale bars represent 500 μm.

Figure 4

flt3 inhibitor recapitulated the effect of flt3 MO knockdown. AC220 (2.5 μmol/L) was used to treat the embryos from 6 hpf to defined developmental stage. Equal volume of DMSO treatment was used as control. (A-B) WISH comparing the l-plastin expression in the anterior yolk sac region between DMSO control (A) and AC220 (B) embryos at 19 hpf. (C) Comparison of l-plastin expression in DMSO control and AC220 embryos at 19 hpf. (D-I) WISH comparing the expression of c-myb (D-E), flt4 (F-G), and efnb2α (H-I) between DMSO control (D,F,H) and AC220 (E,G,I) embryos at 32 hpf. The arrowheads in panel D indicated c-myb expression along the ventral wall of DA. (J-M) WISH showing the expression of c-myb (J-K) and rag1 (L-M) expression in DMSO control (J,L) and AC220 (K,M) embryos at 96 hpf. The arrow and arrowheads in panel J indicated c-myb expression at the thymus and CHT. The circled region (L-M) indicates the rag1 expression in the thymus. Scale bars represent 500 μm.

Close modal

FLT3-ITD overexpression significantly induced ectopic myeloid cell expansion during early embryogenesis

We first cloned the human FLT3-ITD into an egfp plasmid, and FLT3-ITD and egfp were separated by a T2a self-cleaving peptide. The FLT3-ITD sequence was confirmed by DNA sequencing (supplemental Figure 8). The FLT3ITD-T2a-egfp plasmid was linearized and microinjected into 1-cell stage embryos (100 pg per embryo). FLT3ITD-T2a-egfp expressed in a mosaic pattern (supplemental Figure 9A-D) with evidence of egfp+ cells could be identified in the anterior yolk sac (supplemental Figure 9E-F) and the CHT (supplemental Figure 9G-H).

Notably, FLT3-ITD expression was associated with a significant expansion of the primitive myeloid cells that were characterized by the expression of pu.1, mpo, cebpα, and l-plastin at 18 (supplemental Figure 10A-H) and 36 hpf (Figure 5A-D). The responses based on WISH were defined arbitrarily as intermediate, in which increased gene expression was identified but no clustering was found, and severe, in which significant clustering of gene expression could be identified on the surface of yolk (supplemental Figures 11-12). FLT3-ITD expression based on the FLT3-ITD-T2a-mRFP plasmid also significantly increased egfp+ population in dissociated Tg(mpo:egfp) embryos at 36 hpf (Figure 5E-F) from a baseline of 0.40% ± 0.27% to 0.57% ± 0.29% (P = .038). The myeloid expansion appeared to be less than that in WISH, as the expression of FLT3-ITD-mRFP was less efficient than that of FLT-ITD-EGFP. Quantification of double mpo:egfp+ and mRFP populations was not performed due to the small populations of cells detected. To further examine the morphology of the myeloid cell expansion, egfp+ cells were isolated by fluorescence-activated cell sorter (FACS). In the latter experiments, mRNA (supplemental Figure 13A-B) was used instead of plasmid DNA as it gave rise to a slightly stronger myeloid expansion in embryos that would facilitate isolation of egfp+ cells (supplemental Figure 13C-D). The sorted egfp+ cells had a monocytic morphology, consistent with its myeloid nature (Figure 5G-J).

Figure 5

FLT3-ITD overexpression induced ectopic myeloid expansion in zebrafish. (A-D) Human WT FLT3 (FLT3-WT) and FLT3-ITD mutation (FLT3-ITD) was cloned into pegfp-N3 vector. The vector containing the FLT3WT-T2a-egfp and FLT3ITD-T2a-egfp transgene was microinjected into 1-cell–stage zebrafish embryos. WISH comparing the pu.1 (A), mpo (B), cebpα (C), and l-plastin (D) expression between uninjected, FLT3-WT, and FLT3-ITD overexpressing embryos at 36 hpf. The arrowheads and arrows indicated the typical intermediate and severe expansion (for definition, see supplemental Figure 11). Panel Ai-iii represents the pu.1 expression in uninjected (Ai), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Bi-iii represents the mpo expression in uninjected (Bi), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Ci-iii represents the cebpα expression in uninjected (Ci), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Di-iii represents the l-plastin expression in uninjected (Di), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. (E-F) Human FLT3-ITD was cloned into the pDsRed-Monomer-N1 vector to generate the FLT3-ITD-T2a-mRFP transgene. The CMV-driven FLT3-ITD-T2a-mRFP transgene was microinjected into 1-cell–stage embryos. The level of increase in mpo+ cells might appear lower as the FLT3-ITD-T2a-mRFP transgene expressed weaker FLT3-ITD than the FLT3ITD-T2a-egfp transgene used for WISH. (G-H) Human FLT3-ITD mRNA was in vitro transcribed and microinjected into 1-cell–stage Tg(mpo:egfp) embryos. The egfp+ cells indicate the mpo expression in uninjected (Gi-ii) and FLT3-ITD mRNA (Hi-ii) embryos at 36 hpf. (I-J) FACS of mpo+ cells by egfp, and the morphology of the mpo+ cells in uninjected (I) and FLT3-ITD mRNA (J) injected embryos were examined (Shandon Cytospin 4; Thermo Electron Corporation) with Wright-Giemsa staining. The mpo:egfp+ cells were sorted (MoFlo XDP; Beckman Coulter) Scale bars represent 500 μm in panels A-D, and ×600 magnification in panels I-J.

Figure 5

FLT3-ITD overexpression induced ectopic myeloid expansion in zebrafish. (A-D) Human WT FLT3 (FLT3-WT) and FLT3-ITD mutation (FLT3-ITD) was cloned into pegfp-N3 vector. The vector containing the FLT3WT-T2a-egfp and FLT3ITD-T2a-egfp transgene was microinjected into 1-cell–stage zebrafish embryos. WISH comparing the pu.1 (A), mpo (B), cebpα (C), and l-plastin (D) expression between uninjected, FLT3-WT, and FLT3-ITD overexpressing embryos at 36 hpf. The arrowheads and arrows indicated the typical intermediate and severe expansion (for definition, see supplemental Figure 11). Panel Ai-iii represents the pu.1 expression in uninjected (Ai), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Bi-iii represents the mpo expression in uninjected (Bi), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Ci-iii represents the cebpα expression in uninjected (Ci), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. Panel Di-iii represents the l-plastin expression in uninjected (Di), FLT3-WT (ii), and FLT3-ITD (iii) embryos, respectively. (E-F) Human FLT3-ITD was cloned into the pDsRed-Monomer-N1 vector to generate the FLT3-ITD-T2a-mRFP transgene. The CMV-driven FLT3-ITD-T2a-mRFP transgene was microinjected into 1-cell–stage embryos. The level of increase in mpo+ cells might appear lower as the FLT3-ITD-T2a-mRFP transgene expressed weaker FLT3-ITD than the FLT3ITD-T2a-egfp transgene used for WISH. (G-H) Human FLT3-ITD mRNA was in vitro transcribed and microinjected into 1-cell–stage Tg(mpo:egfp) embryos. The egfp+ cells indicate the mpo expression in uninjected (Gi-ii) and FLT3-ITD mRNA (Hi-ii) embryos at 36 hpf. (I-J) FACS of mpo+ cells by egfp, and the morphology of the mpo+ cells in uninjected (I) and FLT3-ITD mRNA (J) injected embryos were examined (Shandon Cytospin 4; Thermo Electron Corporation) with Wright-Giemsa staining. The mpo:egfp+ cells were sorted (MoFlo XDP; Beckman Coulter) Scale bars represent 500 μm in panels A-D, and ×600 magnification in panels I-J.

Close modal

Experiments were also performed to examine whether the myeloid expansion resulted from an increased flt3 signaling in the embryos. Overexpression of human FLT3-ITD in embryos was also associated with an increase in p-flt3, p-stat5, p-erk1/2, and p-akt at 36 hpf (Figure 6A). In addition, colocalization of mpo and egfp (surrogate for FLT3-ITD) was evident (Figure 6B-E). Furthermore, the effects of AC220 treatment on myeloid responses to FLT3-ITD were evaluated. AC220 had no effect on baseline pu.1+, mpo+, and cebpα+ populations in uninjected embryos (Figure 7A-F). However, it significantly ameliorated the myeloid expansion induced by FLT3-ITD expression based on pu.1+, mpo+, and cebpα+ expression (Figure 7G-L,S-U, P < .01 in all cases, χ2 test, N = 3 experiments). Collectively, these observations supported the proposition that human FLT3-ITD expression stimulated endogenous flt3 signaling in zebrafish and led to expansion of myeloid populations. The possibility of aberrant sequestration of these myeloid populations in the Duct of Cuvier was unlikely as the circulatory system was intact based on direct visualization (supplemental Video 1) and myeloid expansion was also evident at 23 hpf, before the onset of circulation (supplemental Figure 10I-P).

Figure 6

FLT3-ITD expression induced evident p-stat5, p-erk, and p-akt, and colocalized with mpo. (A) Western blotting comparing the FLT3, p-FLT3, stat5, p-stat5, erk1/2, p-erk1/2, akt, and p-akt in uninjected, FLT3-WT, and FLT3-ITD embryos at 36 hpf. (B-E) Double-color WISH showing the mpo and egfp expression in uninjected control (B-C) and FLT3-ITD overexpressing (D-E) embryos at 36 hpf. The boxed area indicates the coexpression of mpo and egfp in the yolk sac. Scale bars represent 500 μm.

Figure 6

FLT3-ITD expression induced evident p-stat5, p-erk, and p-akt, and colocalized with mpo. (A) Western blotting comparing the FLT3, p-FLT3, stat5, p-stat5, erk1/2, p-erk1/2, akt, and p-akt in uninjected, FLT3-WT, and FLT3-ITD embryos at 36 hpf. (B-E) Double-color WISH showing the mpo and egfp expression in uninjected control (B-C) and FLT3-ITD overexpressing (D-E) embryos at 36 hpf. The boxed area indicates the coexpression of mpo and egfp in the yolk sac. Scale bars represent 500 μm.

Close modal
Figure 7

AC220 effectively ameliorated FLT3-ITD, but not FLT3-TKD–induced, myeloid expansion. The human D835Y TKD mutation (FLT3-TKD) was generated based on the vector containing the human FLT3-WT sequence by a site-directed mutagenesis. The vectors containing the FLT3-ITD-T2a-egfp and FLT3-TKD-T2a-egfp were microinjected into 1-cell–stage embryos. Embryos were treated with DMSO control or AC220 (2.5 μmol/L) from 6 hpf to 36 hpf. (A-F) WISH showing the expression of pu.1 (A-B), mpo (C-D), and cebpα (E-F) in the uninjected embryos treated with DMSO and AC220 from 6 to 36 hpf, respectively. (G-L) WISH showing the expression of pu.1 (G-H), mpo (I-J), and cebpα (K-L) in the FLT3-ITD embryos treated with DMSO and AC220 from 6 to 36 hpf. (M-R) WISH showing pu.1 (M-N), mpo (O-P), and cebpα (Q-R) expression in the FLT3-TKD embryos treated with DMSO and AC220 treatment from 6 to 36 hpf. (S-U) Percentage of FLT3-ITD, FLT3-TKD embryos showing the myeloid-committed cell expansion as shown by pu.1 (S), mpo (T), and cebpα (U) expansion after DMSO or AC220 treatment from 6 to 36 hpf. AC220 significantly ameliorated the expansion of myeloid expression based on pu.1, mpo, and cebpα expression (P < .01) in FLT3-ITD but not FLT3-TKD embryos (P > .05). Scale bars represent 500 μm.

Figure 7

AC220 effectively ameliorated FLT3-ITD, but not FLT3-TKD–induced, myeloid expansion. The human D835Y TKD mutation (FLT3-TKD) was generated based on the vector containing the human FLT3-WT sequence by a site-directed mutagenesis. The vectors containing the FLT3-ITD-T2a-egfp and FLT3-TKD-T2a-egfp were microinjected into 1-cell–stage embryos. Embryos were treated with DMSO control or AC220 (2.5 μmol/L) from 6 hpf to 36 hpf. (A-F) WISH showing the expression of pu.1 (A-B), mpo (C-D), and cebpα (E-F) in the uninjected embryos treated with DMSO and AC220 from 6 to 36 hpf, respectively. (G-L) WISH showing the expression of pu.1 (G-H), mpo (I-J), and cebpα (K-L) in the FLT3-ITD embryos treated with DMSO and AC220 from 6 to 36 hpf. (M-R) WISH showing pu.1 (M-N), mpo (O-P), and cebpα (Q-R) expression in the FLT3-TKD embryos treated with DMSO and AC220 treatment from 6 to 36 hpf. (S-U) Percentage of FLT3-ITD, FLT3-TKD embryos showing the myeloid-committed cell expansion as shown by pu.1 (S), mpo (T), and cebpα (U) expansion after DMSO or AC220 treatment from 6 to 36 hpf. AC220 significantly ameliorated the expansion of myeloid expression based on pu.1, mpo, and cebpα expression (P < .01) in FLT3-ITD but not FLT3-TKD embryos (P > .05). Scale bars represent 500 μm.

Close modal

FLT3-TKD (D835Y) overexpression induced ectopic myeloid cell expansion resistant to AC220

Single-base mutation of the FLT3-TKD occurs in ∼5% AML and its prognostic significance is unclear.22  In zebrafish embryos, overexpression of FLT3-TKD (D835Y) likewise induced a significant expansion of pu.1 (Figure 7M), mpo (Figure 7O), and cebpα (Figure 7Q) despite a smaller phenotypic penetrance for mpo and cebpα. Importantly, AC220 had no significant ameliorating effects on myeloid expansion in the FLT3-TKD embryos (Figure 7N,P,R-U, P > .05 in all gene expression studies, χ2 test).

Overexpression of FLT3-ITD-TKD double mutation in zebrafish embryos conferred resistance to AC220 treatment

Both clinical and laboratory data demonstrated that the leukemia clone carrying the additional TKD mutation might emerge in FLT3-ITD+ AML treated with TKI (the “double mutation”), leading to drug resistance.26,27  We addressed this issue in zebrafish embryos by injecting them with plasmid encoding the FLT3-ITD-TKD (D835Y) double mutation. Overexpression of the double mutants induced expansion of myeloid compartment as shown by the increase in pu.1, mpo, cebpα, and l-plastin expression (Figure 8E-H). However, AC220 did not significantly reduce the expansion of the myeloid compartment (Figure 8I-L and M-P, P > .05 in all gene expression studies, χ2 test), suggesting that the double mutation has conferred resistance to AC220 in zebrafish embryos.

Figure 8

FLT3-ITD-TKD double mutation showed poor response to AC220 comparing to FLT3-ITD. The human FLT3-ITD-TKD double mutation was generated by inducing TKD mutation into the FLT3-ITD sequence using the site-directed mutagenesis approach. WISH showing normal expression of pu.1 (A,E,I), mpo (B,F,J), cebpα (C,G,K), l-plastin (D,H,L) in uninjected control (A-D), FLT3-ITD-TKD embryos treated with DMSO (E-H) and AC220 (I-L) from 6 to 36 hpf. (M-P) Comparison of pu.1 (M), mpo (N), cebpα (O), and l-plastin (P) expansion in FLT3-ITD-TKD embryos treated with DMSO or AC220 from 6 to 36 hpf. There was no statistically significant effect of AC220 treatment (P > .05). Scale bars represent 500 μm.

Figure 8

FLT3-ITD-TKD double mutation showed poor response to AC220 comparing to FLT3-ITD. The human FLT3-ITD-TKD double mutation was generated by inducing TKD mutation into the FLT3-ITD sequence using the site-directed mutagenesis approach. WISH showing normal expression of pu.1 (A,E,I), mpo (B,F,J), cebpα (C,G,K), l-plastin (D,H,L) in uninjected control (A-D), FLT3-ITD-TKD embryos treated with DMSO (E-H) and AC220 (I-L) from 6 to 36 hpf. (M-P) Comparison of pu.1 (M), mpo (N), cebpα (O), and l-plastin (P) expansion in FLT3-ITD-TKD embryos treated with DMSO or AC220 from 6 to 36 hpf. There was no statistically significant effect of AC220 treatment (P > .05). Scale bars represent 500 μm.

Close modal

In the present study, we demonstrated that flt3 was highly conserved at the JMD and TKD and its targeting by MO and AC220 specifically reduced primitive myelopoiesis and definitive HSPC specification. The effects of knockdown on primitive myelopoiesis were seen as early as 18 hpf, with a preferential downregulation of l-plastin but not pu.1 expression. csf1r and mpeg1 expression at 22 hpf, before the onset of circulating, were significantly reduced, suggesting flt3 may play an important role in macrophage development during primitive myelopoiesis. Proliferation of primitive macrophages was reduced in the flt3 morphants as shown by phospho-Histone H3 staining in mpeg1+ cells. Apoptosis was not affected. Primitive erythroid and precursor cells were not affected, supporting the proposition that flt3 knockdown perturbed specifically macrophage development rather than erythromyeloid lineage specification. The effects on c-myb and cd41 populations in the ventral wall of DA as well as the lck and rag-1 populations in the thymus might reflect perturbation of the hematopoietic progenitor population. The results were consistent with the expression of flt3 in the ventral wall of DA and PBI. As the specification of hematopoietic stem and progenitors depends on blood flow through the axial circulation,42  we also evaluated the process of vasculogenesis in the morphant embryos. Vasculogenesis and arteriovenous specification as well as the blood flow in the vasculature of the morphants (supplemental Videos 2-5) were intact, suggesting that flt3 might specify definitive HSPC after its derivation from the putative hemogenic endothelium.

We have also evaluated the effects of FLT3 activation during embryonic development. Intriguingly, expression of human FLT3-ITD was able to activate downstream signaling in zebrafish embryos, highlighting the conserved signaling machinery in response to FLT3 activation in this model. Primitive myelopoiesis was accentuated, as shown by the increased populations of mpo, cebpα, l-plastin, and pu.1 positive cells. Microinjection of mRNA encoding for FLT3-ITD, which enabled earlier and more ubiquitous FLT3-ITD expression, only affected myeloid development but not primitive precursors and erythropoiesis (supplemental Figure 14). Therefore, FLT3-ITD overexpression might induce proliferation of primitive myeloid cells rather than misspecification or skewing of lineage differentiation. Colocalization of FLT3-ITD expression and mpo could be demonstrated, supporting the proposition that constitutive activation of FLT3 might be driving the expansion of the myeloid compartment. The ectopic myeloid expansion corroborated with murine studies in which Flt3 knockin induced myeloid expansion reminiscent of human myeloproliferative neoplasms44  and AML with short latency in the presence of cooperative oncogenic signaling.45,46  Importantly, coexisting TKD in the same allele conferred the myeloid compartment with the resistance to AC220, consistent with the clonal emergence of AC220-resistant AML patients.

The results of this study provided an example whereby human AML can be modeled in this organism. To demonstrate the clinical relevance of this model, we made use of AC220 that has been tested in clinical trials for the treatment of AML. Interestingly, the myeloid expansion induced by TKD mutant expression was resistant to AC220, consistent with the clinical emergence of resistant clones carrying additional TKD at D835 position. Off-target effects of multikinase inhibitors could be readily demonstrated, as exemplified by the perturbation on arterial specification upon sorafenib and sunitinib treatment secondary to disruption of vascular endothelial growth factor (VEGF) signaling (supplemental Figure 15).

We envisioned that the model might be modified to suit specific clinical needs so that novel and patient-specific therapeutic agents can be identified at a whole-organism level in real time. For instance, multiple FLT3-ITD clones that often coexist in AML samples can be isolated at different stages of treatment and expressed in zebrafish embryos and the inhibitory effects of TKI on different ITD clones can be evaluated. The efficacies of novel TKI and inhibitors of downstream signals including pSTAT5,23  pAKT,24  and PIM147  toward resistant FLT3-ITD clones could also be evaluated. In addition to FLT3-ITD, other mutant leukemia genes have been evaluated. For instance, the mutant human nucleophosmin 1 (NPM1c) gene, when expressed in zebrafish embryos, has been shown to induce myeloid expansion with the mutant protein resided exclusively in the cytoplasm, reminiscent of human NPM1c+ AML.48  With the advent of next-generation sequencing and the emergence of new mutations in AML and related diseases,49  the zebrafish may provide a timely model whereby the pathogenetic significance and therapeutic potential of novel gene mutations can be evaluated.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Dr Leonard Zon for the helpful comments on the data and Dr Zilong Wen for the procurement of embryos.

This work was supported by the General Research Fund (HKU771110 and HKU729809M), the Innovative Collaborative Research Programme and Zebrafish Core Facility at the Li Ka Shing Faculty of Medicine, as well as National Institutes of Health (General Medical Sciences) grant GM63904 (S.C.E.).

A.Y.H.L. is the Li Shu Fan Medical Foundation Professor in Haematology and received funding from the endowment.

Contribution: B.-L.H. designed and performed the experiments, analyzed the data, and wrote and approved the manuscript; C.H.M., X.S., and H.C.H.C. performed the cloning, western blot, and microinjection experiments and approved the manuscript; A.C.H.M. and S.C.E. designed the experiments, analyzed the data, and approved the manuscript; C.W.E.S. cloned the human FLT3-ITD and approved the manuscript; W.W.L.C. performed the cell staining and evaluated cellular morphology and approved the manuscript; W.Z. and Y.Z. prepared the cloche mutant embryos and approved the manuscript; and A.Y.H.L. designed the experiments, analyzed the data, and wrote and approved the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Anskar Y. H. Leung, Department of Medicine, Queen Mary Hospital, Room K418, K Block, Pok Fu Lam Rd, Pokfulam, Hong Kong; e-mail: ayhleung@hku.hk.

1
Davidson
 
AJ
Zon
 
LI
The ‘definitive’ (and ‘primitive’) guide to zebrafish hematopoiesis.
Oncogene
2004
, vol. 
23
 
43
(pg. 
7233
-
7246
)
2
Xu
 
J
Du
 
L
Wen
 
Z
Myelopoiesis during zebrafish early development.
J Genet Genomics
2012
, vol. 
39
 
9
(pg. 
435
-
442
)
3
Lieschke
 
GJ
Oates
 
AC
Paw
 
BH
, et al. 
Zebrafish SPI-1 (PU.1) marks a site of myeloid development independent of primitive erythropoiesis: implications for axial patterning.
Dev Biol
2002
, vol. 
246
 
2
(pg. 
274
-
295
)
4
Ellett
 
F
Lieschke
 
GJ
Zebrafish as a model for vertebrate hematopoiesis.
Curr Opin Pharmacol
2010
, vol. 
10
 
5
(pg. 
563
-
570
)
5
Meijer
 
AH
van der Sar
 
AM
Cunha
 
C
, et al. 
Identification and real-time imaging of a myc-expressing neutrophil population involved in inflammation and mycobacterial granuloma formation in zebrafish.
Dev Comp Immunol
2008
, vol. 
32
 
1
(pg. 
36
-
49
)
6
Hall
 
C
Flores
 
MV
Storm
 
T
Crosier
 
K
Crosier
 
P
The zebrafish lysozyme C promoter drives myeloid-specific expression in transgenic fish.
BMC Dev Biol
2007
, vol. 
7
 pg. 
42
 
7
Le Guyader
 
D
Redd
 
MJ
Colucci-Guyon
 
E
, et al. 
Origins and unconventional behavior of neutrophils in developing zebrafish.
Blood
2008
, vol. 
111
 
1
(pg. 
132
-
141
)
8
Zakrzewska
 
A
Cui
 
C
Stockhammer
 
OW
Benard
 
EL
Spaink
 
HP
Meijer
 
AH
Macrophage-specific gene functions in Spi1-directed innate immunity.
Blood
2010
, vol. 
116
 
3
(pg. 
e1
-
e11
)
9
Bertrand
 
JY
Kim
 
AD
Violette
 
EP
Stachura
 
DL
Cisson
 
JL
Traver
 
D
Definitive hematopoiesis initiates through a committed erythromyeloid progenitor in the zebrafish embryo.
Development
2007
, vol. 
134
 
23
(pg. 
4147
-
4156
)
10
Zhang
 
Y
Jin
 
H
Li
 
L
Qin
 
FX
Wen
 
Z
cMyb regulates hematopoietic stem/progenitor cell mobilization during zebrafish hematopoiesis.
Blood
2011
, vol. 
118
 
15
(pg. 
4093
-
4101
)
11
de Jong
 
JL
Zon
 
LI
Use of the zebrafish system to study primitive and definitive hematopoiesis.
Annu Rev Genet
2005
, vol. 
39
 (pg. 
481
-
501
)
12
Rhodes
 
J
Hagen
 
A
Hsu
 
K
, et al. 
Interplay of pu.1 and gata1 determines myelo-erythroid progenitor cell fate in zebrafish.
Dev Cell
2005
, vol. 
8
 
1
(pg. 
97
-
108
)
13
Galloway
 
JL
Wingert
 
RA
Thisse
 
C
Thisse
 
B
Zon
 
LI
Loss of gata1 but not gata2 converts erythropoiesis to myelopoiesis in zebrafish embryos.
Dev Cell
2005
, vol. 
8
 
1
(pg. 
109
-
116
)
14
Yuan
 
H
Zhou
 
J
Deng
 
M
, et al. 
Sumoylation of CCAAT/enhancer-binding protein α promotes the biased primitive hematopoiesis of zebrafish.
Blood
2011
, vol. 
117
 
26
(pg. 
7014
-
7020
)
15
Small
 
D
Levenstein
 
M
Kim
 
E
, et al. 
STK-1, the human homolog of Flk-2/Flt-3, is selectively expressed in CD34+ human bone marrow cells and is involved in the proliferation of early progenitor/stem cells.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 
2
(pg. 
459
-
463
)
16
Stirewalt
 
DL
Radich
 
JP
The role of FLT3 in haematopoietic malignancies.
Nat Rev Cancer
2003
, vol. 
3
 
9
(pg. 
650
-
665
)
17
Rosnet
 
O
Schiff
 
C
Pébusque
 
MJ
, et al. 
Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells.
Blood
1993
, vol. 
82
 
4
(pg. 
1110
-
1119
)
18
Matthews
 
W
Jordan
 
CT
Wiegand
 
GW
Pardoll
 
D
Lemischka
 
IR
A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations.
Cell
1991
, vol. 
65
 
7
(pg. 
1143
-
1152
)
19
Ueda
 
T
Tsuji
 
K
Yoshino
 
H
, et al. 
Expansion of human NOD/SCID-repopulating cells by stem cell factor, Flk2/Flt3 ligand, thrombopoietin, IL-6, and soluble IL-6 receptor.
J Clin Invest
2000
, vol. 
105
 
7
(pg. 
1013
-
1021
)
20
Mackarehtschian
 
K
Hardin
 
JD
Moore
 
KA
Boast
 
S
Goff
 
SP
Lemischka
 
IR
Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors.
Immunity
1995
, vol. 
3
 
1
(pg. 
147
-
161
)
21
Buza-Vidas
 
N
Cheng
 
M
Duarte
 
S
Charoudeh
 
HN
Jacobsen
 
SE
Sitnicka
 
E
FLT3 receptor and ligand are dispensable for maintenance and posttransplantation expansion of mouse hematopoietic stem cells.
Blood
2009
, vol. 
113
 
15
(pg. 
3453
-
3460
)
22
Zeisig
 
BB
Kulasekararaj
 
AG
Mufti
 
GJ
So
 
CW
SnapShot: acute myeloid leukemia.
Cancer Cell
2012
 
22(5):698-698.e1
23
Choudhary
 
C
Brandts
 
C
Schwable
 
J
, et al. 
Activation mechanisms of STAT5 by oncogenic Flt3-ITD.
Blood
2007
, vol. 
110
 
1
(pg. 
370
-
374
)
24
Brandts
 
CH
Sargin
 
B
Rode
 
M
, et al. 
Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation.
Cancer Res
2005
, vol. 
65
 
21
(pg. 
9643
-
9650
)
25
Nishioka
 
C
Ikezoe
 
T
Yang
 
J
, et al. 
Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene.
Leuk Res
2008
, vol. 
32
 
6
(pg. 
865
-
872
)
26
Man
 
CH
Fung
 
TK
Ho
 
C
, et al. 
Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation.
Blood
2012
, vol. 
119
 
22
(pg. 
5133
-
5143
)
27
Smith
 
CC
Wang
 
Q
Chin
 
CS
, et al. 
Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia.
Nature
2012
, vol. 
485
 
7397
(pg. 
260
-
263
)
28
Leung
 
AY
Man
 
CH
Kwong
 
YL
FLT3 inhibition: a moving and evolving target in acute myeloid leukaemia.
Leukemia
2013
, vol. 
27
 
2
(pg. 
260
-
268
)
29
Ma
 
AC
Ward
 
AC
Liang
 
R
Leung
 
AY
The role of jak2a in zebrafish hematopoiesis.
Blood
2007
, vol. 
110
 
6
(pg. 
1824
-
1830
)
30
Kimmel
 
CB
Ballard
 
WW
Kimmel
 
SR
Ullmann
 
B
Schilling
 
TF
Stages of embryonic development of the zebrafish.
Dev Dyn
1995
, vol. 
203
 
3
(pg. 
253
-
310
)
31
Nasevicius
 
A
Ekker
 
SC
Effective targeted gene ‘knockdown’ in zebrafish.
Nat Genet
2000
, vol. 
26
 
2
(pg. 
216
-
220
)
32
Ekker
 
SC
Morphants: a new systematic vertebrate functional genomics approach.
Yeast
2000
, vol. 
17
 
4
(pg. 
302
-
306
)
33
Thisse
 
C
Thisse
 
B
High-resolution in situ hybridization to whole-mount zebrafish embryos.
Nat Protoc
2008
, vol. 
3
 
1
(pg. 
59
-
69
)
34
Liang
 
D
Chang
 
JR
Chin
 
AJ
, et al. 
The role of vascular endothelial growth factor (VEGF) in vasculogenesis, angiogenesis, and hematopoiesis in zebrafish development.
Mech Dev
2001
, vol. 
108
 
1-2
(pg. 
29
-
43
)
35
Ma
 
AC
Chung
 
MI
Liang
 
R
Leung
 
AY
The role of survivin2 in primitive hematopoiesis during zebrafish development.
Leukemia
2009
, vol. 
23
 
4
(pg. 
712
-
720
)
36
Tang
 
R
Dodd
 
A
Lai
 
D
McNabb
 
WC
Love
 
DR
Validation of zebrafish (Danio rerio) reference genes for quantitative real-time RT-PCR normalization.
Acta Biochim Biophys Sin (Shanghai)
2007
, vol. 
39
 
5
(pg. 
384
-
390
)
37
Ji
 
W
Zhou
 
W
Abruzzese
 
R
, et al. 
A method for determining zygosity of transgenic zebrafish by TaqMan real-time PCR.
Anal Biochem
2005
, vol. 
344
 
2
(pg. 
240
-
246
)
38
Choorapoikayil
 
S
Kers
 
R
Herbomel
 
P
Kissa
 
K
den Hertog
 
J
Pivotal role of Pten in the balance between proliferation and differentiation of hematopoietic stem cells in zebrafish.
Blood
2014
, vol. 
123
 
2
(pg. 
184
-
190
)
39
Wilhelm
 
S
Carter
 
C
Lynch
 
M
, et al. 
Discovery and development of sorafenib: a multikinase inhibitor for treating cancer.
Nat Rev Drug Discov
2006
, vol. 
5
 
10
(pg. 
835
-
844
)
40
O’Farrell
 
AM
Abrams
 
TJ
Yuen
 
HA
, et al. 
SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo.
Blood
2003
, vol. 
101
 
9
(pg. 
3597
-
3605
)
41
Zarrinkar
 
PP
Gunawardane
 
RN
Cramer
 
MD
, et al. 
AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML).
Blood
2009
, vol. 
114
 
14
(pg. 
2984
-
2992
)
42
North
 
TE
Goessling
 
W
Peeters
 
M
, et al. 
Hematopoietic stem cell development is dependent on blood flow.
Cell
2009
, vol. 
137
 
4
(pg. 
736
-
748
)
43
Taylor
 
SJ
Dagger
 
SA
Thien
 
CB
Wikstrom
 
ME
Langdon
 
WY
Flt3 inhibitor AC220 is a potent therapy in a mouse model of myeloproliferative disease driven by enhanced wild-type Flt3 signaling.
Blood
2012
, vol. 
120
 
19
(pg. 
4049
-
4057
)
44
Li
 
L
Piloto
 
O
Nguyen
 
HB
, et al. 
Knock-in of an internal tandem duplication mutation into murine FLT3 confers myeloproliferative disease in a mouse model.
Blood
2008
, vol. 
111
 
7
(pg. 
3849
-
3858
)
45
Zorko
 
NA
Bernot
 
KM
Whitman
 
SP
, et al. 
Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias.
Blood
2012
, vol. 
120
 
5
(pg. 
1130
-
1136
)
46
Greenblatt
 
S
Li
 
L
Slape
 
C
, et al. 
Knock-in of a FLT3/ITD mutation cooperates with a NUP98-HOXD13 fusion to generate acute myeloid leukemia in a mouse model.
Blood
2012
, vol. 
119
 
12
(pg. 
2883
-
2894
)
47
Fathi
 
AT
Arowojolu
 
O
Swinnen
 
I
, et al. 
A potential therapeutic target for FLT3-ITD AML: PIM1 kinase.
Leuk Res
2012
, vol. 
36
 
2
(pg. 
224
-
231
)
48
Bolli
 
N
Payne
 
EM
Grabher
 
C
, et al. 
Expression of the cytoplasmic NPM1 mutant (NPMc+) causes the expansion of hematopoietic cells in zebrafish.
Blood
2010
, vol. 
115
 
16
(pg. 
3329
-
3340
)
49
Patel
 
JP
Gönen
 
M
Figueroa
 
ME
, et al. 
Prognostic relevance of integrated genetic profiling in acute myeloid leukemia.
N Engl J Med
2012
, vol. 
366
 
12
(pg. 
1079
-
1089
)
Sign in via your Institution