The interconnected Insulin/IGF signaling (IlS) and Target of Rapamycin (TOR) signaling pathways constitute the main branches of the nutrient-sensing system that couples growth to nutritional conditions in Drosophila. Here, we addressed the influence of these pathways and of diet restriction on the balance between the maintenance of multipotent hematopoietic progenitors and their differentiation in the Drosophila lymph gland. In this larval hematopoietic organ, a pool of stem-like progenitor blood cells (prohemocytes) is kept undifferentiated in response to signaling from a specialized group of cells forming the posterior signaling center (PSC), which serves as a stem cell niche. We show that, reminiscent of the situation in human, loss of the negative regulator of IIS Pten results in lymph gland hyperplasia, aberrant blood cell differentiation and hematopoietic progenitor exhaustion. Using site-directed loss- and gain-of-function analysis, we demonstrate that components of the IIS/TOR pathways control lymph gland homeostasis at two levels. First, they cell-autonomously regulate the size and activity of the hematopoietic niche. Second, they are required within the prohemocytes to control their growth and maintenance. Moreover, we show that diet restriction or genetic alteration mimicking amino acid deprivation triggers progenitor cell differentiation. Hence, our study highlights the role of the IIS/TOR pathways in orchestrating hematopoietic progenitor fate and links blood cell fate to nutritional status.

Thanks to the phylogenetic conservation of the genetic pathways and developmental strategies at stake during blood cell development, Drosophila has emerged as a valuable model organism to study the fundamental mechanisms underlying hematopoiesis (Hartenstein, 2006). Notably, the Drosophila larval hematopoietic organ, the lymph gland, provides a paradigm to reveal the relationships between the hematopoietic niche, the blood cell progenitors and their differentiated progenies (for a review, see Krzemien et al., 2010a). The lymph gland produces three mature blood cell types functionally related to mammalian myeloid cells: the plasmatocytes, the crystal cells and, upon some immune challenges, the lamellocytes (Lanot et al., 2001; Meister and Lagueux, 2003). During the two first instar larval stages, the lymph gland grows and is primarily composed of stem cell-like progenitor blood cells called prohemocytes (Crozatier et al., 2004; Jung et al., 2005; Krzemien et al., 2010b; Minakhina and Steward, 2010). In third instar larvae, the mature lymph gland consists of a pair of primary lobes and several smaller secondary lobes (Fig. 1A). The primary lobes are organized in three zones: the cortical zone (CZ), the medullary zone (MZ) and the posterior signaling center (PSC) (Jung et al., 2005). The MZ as well as the secondary lobes comprise quiescent prohemocytes whereas the CZ contains differentiated hemocytes that emerge from the MZ. Reminiscent of the mammalian hematopoietic system, the balance between stem cell-like and differentiated blood cells is controlled by the PSC, which acts as a niche and secretes diffusible molecules promoting prohemocyte maintenance (Krzemien et al., 2007; Mandal et al., 2007; Sinenko et al., 2009). However, only a handful of signaling pathways controlling niche development or hematopoietic progenitor maintenance are known.

An important challenge for every metazoan is to adjust its development to food availability, and growing evidence indicates that nutrition can influence stem/progenitor cell fate (Amcheslavsky et al., 2009; Chell and Brand, 2010; McLeod et al., 2010; Sousa-Nunes et al., 2011). The interconnected Insulin/IGF signaling (IIS) and Target of Rapamycin (TOR) pathways (Fig. 1B) are the integrators between tissue growth and dietary conditions (Tennessen and Thummel, 2011). Here, we show that these pathways play a crucial role during hematopoiesis in the Drosophila lymph gland. First, they regulate the size and activity of the hematopoietic niche. Second, they cell-autonomously control blood cell progenitor maintenance. Finally, they couple blood cell development to the larval nutritional status.

Drosophila strains and larvae collections

The following fly stocks were used: Pcol85-Gal4, UAS::mCD8-GFP (Krzemien et al., 2007) referred here as col-Gal4, ppl-Gal4/CyOGal80; slifAnti, ppl-Gal4/CyO (Colombani et al., 2003), PTEN100/CyO Act-GFP, PTEN117/CyO Act-GFP (Oldham et al., 2002), UAS::TSC1, UAS::TSC2 (Tapon et al., 2001), UAS::PTEN (Huang et al., 1999), UAS::PI3Kcaax/Cyo (Leevers et al., 1996), UAS::dFoxo (Puig et al., 2003), tepIV-Gal4 (Avet-Rochex et al., 2010), Cg25C-GFP and Bc-GFP (Sorrentino et al., 2007), Hh-GFP (Tokusumi et al., 2011). The RNAi stocks were obtained from Vienna Drosophila RNAi Center (VDRC).

Embryos were collected for 6 hours and equivalent numbers of hatching first instar larvae were transferred to vials with regular food medium at 25°C until wandering third instar larval stage. Starvation experiments were performed as established by Ikeya et al. (Ikeya et al., 2002). Larvae from crosses of UAS or Gal4 lines to w1118 were used as controls.

Fig. 1.

IIS signaling controls lymph gland hematopoiesis. (A) Schematic of the Drosophila third instar lymph gland. The hematopoietic niche (green), blood cell progenitors (yellow) and mature blood cells (red) are depicted. Primary (prim) and secondary (sec) lobes are separated by pericardiac cells (black, PP). (B) Schematic of the IIS/TOR pathways. (C-J) Third instar Drosophila lymph glands. (C,D) Expression of the prohemocyte marker TepIV. (E,F) Expression of the crystal cell marker Hnt (green) and of the prohemocyte marker Ance (red). (G,H) Immunostaining against phosphorylated Histone H3 (PH3, green) and the plasmatocyte marker P1 (red). Nuclei are labeled with Topro3 (blue). (I,J) Expression of the PSC marker Antp. The PSC is composed of 42 (±14) or 55 (±11) cells in control or Pten100/117 lymph glands, respectively. Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bars: 100 μm.

Fig. 1.

IIS signaling controls lymph gland hematopoiesis. (A) Schematic of the Drosophila third instar lymph gland. The hematopoietic niche (green), blood cell progenitors (yellow) and mature blood cells (red) are depicted. Primary (prim) and secondary (sec) lobes are separated by pericardiac cells (black, PP). (B) Schematic of the IIS/TOR pathways. (C-J) Third instar Drosophila lymph glands. (C,D) Expression of the prohemocyte marker TepIV. (E,F) Expression of the crystal cell marker Hnt (green) and of the prohemocyte marker Ance (red). (G,H) Immunostaining against phosphorylated Histone H3 (PH3, green) and the plasmatocyte marker P1 (red). Nuclei are labeled with Topro3 (blue). (I,J) Expression of the PSC marker Antp. The PSC is composed of 42 (±14) or 55 (±11) cells in control or Pten100/117 lymph glands, respectively. Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bars: 100 μm.

Immunohistochemistry, in situ hybridization and quantification

Lymph glands were processed and stained as described (Avet-Rochex et al., 2010). The following primary antibodies were used: anti-Antp, anti-Hnt (Developmental Studies Hybridoma Bank), anti-Col (Crozatier et al., 2004), anti-P1/NimrodC1 (Vilmos et al., 2004), anti-H3P (Upstate), anti-β-Gal (Cappel), anti-GFP (Torrey Pines). For in situ hybridization, DIG-labeled antisense RNA probes against TepIV, Ance and α-PS4 were used. Samples were visualized with a laser scanning confocal microscope (Leica). Confocal images are displayed as maximum intensity projections of the lymph glands.

For quantification, lymph glands were scanned at an optimized number of slices using a Leica SP5 microscope. Crystal cell and plasmatocyte differentiation indexes were defined as the number of Hnt+ cells (quantified using Volocity software) or the level of Cg25C-GFP expression (measured with Image J software), respectively, in each primary lobe reported to the estimated lobe’s volume. PSC cell number and MZ or lobe size were quantified using Image J software. A minimum of 12 lobes was scored per genotype.

IIS controls lymph gland homeostasis

To test whether the IIS pathway controls lymph gland homeostasis, we used a combination of Pten mutant alleles for which third instar larvae can be obtained (Oldham et al., 2002). Pten codes for a protein phosphatase that antagonizes IIS-induced activation of the Phosphoinositol-3-Kinase (PI3K) (Fig. 1B). Hence, Pten larvae exhibit increased IIS activity. In contrast to wild-type lymph glands, which exhibited strong TepIV and Ance expression in the MZ of their primary lobes and in their secondary lobes, Pten lymph glands displayed markedly reduced expression of these two prohemocyte markers (Fig. 1C-F; supplementary material Fig. S1). Conversely, immunostaining against the crystal cell marker Hindsight (Hnt; Peb – FlyBase) (supplementary material Fig. S1) or the plasmatocyte marker P1, showed that whereas differentiated hemocytes are normally restricted to the CZ, they filled Pten lymph glands primary lobes and were also present in the secondary lobes (Fig. 1E-H), which is a characteristic sign of precocious differentiation (Owusu-Ansah and Banerjee, 2009). In addition, in situ hybridization against α-PS4 revealed the presence of lamellocytes, which are seldom observed in the normal situation (supplementary material Fig. S2). Furthermore, Pten lymph glands frequently exhibited premature primary lobe dispersal and displayed overgrown primary and secondary lobes (supplementary material Fig. S5). Consistent with this observation, anti-phospho-H3 labeling showed that Pten lymph glands contained numerous proliferating cells (Fig. 1G,H). Thus, as in mammals, in which Pten loss induces hematopoietic stem cell exhaustion and myeloproliferative disease (Yilmaz et al., 2006; Zhang et al., 2006), Pten is required to prevent the aberrant differentiation of the blood cell progenitors into the three mature blood cell types found in Drosophila.

To ensure that the massive differentiation observed in Pten mutants was not due to a defect in PSC cell specification, we monitored the expression of the two key regulators of PSC fate: Antennapedia (Antp) and Collier (Col; Kn – FlyBase) (Krzemien et al., 2007; Mandal et al., 2007). Although immunostaining against these two proteins demonstrated that PSC cells were correctly specified, we found that the size of the niche was strongly increased (Fig. 1I,J; data not shown), suggesting that IIS normally controls PSC cell number. As the presence of a larger niche was nonetheless associated with increased prohemocyte differentiation, we surmised that these contradictory phenotypes might arise from a dual activity of the IIS in the PSC versus the MZ. We thus assessed its role specifically in these two domains.

IIS and TOR pathways cell-autonomously control the size of the hematopoietic niche

To test whether the IIS pathway is cell-autonomously required in the PSC, we used the col-Gal4 driver (Krzemien et al., 2007), expression of which is strictly confined to the PSC during lymph gland ontogeny, as demonstrated by a lineage tracing experiment (supplementary material Fig. S3). In addition, we took advantage of col-Gal4-driven expression in the wing disc to confirm the specificity of the UAS transgenes used in this study (supplementary material Fig. S4). As observed in Pten larvae, over-activation of IIS in the PSC, induced by expressing either Pten RNAi or an active form of PI3K (PI3Kcaax) (Leevers et al., 1996), led to a strong increase in PSC size (Fig. 2B,C). This phenotype correlated with a rise in PSC cell number (Fig. 2G). Conversely, knocking down InR by RNAi or overexpressing Pten caused a reduction in PSC cell number (Fig. 2D,G). As IIS impinges on TOR activity (Fig. 1B), we tested whether this pathway also regulates PSC development. PSC cell number diminished when the TOR pathway was inactivated either by overexpressing both TSC1 and TSC2 (gig – FlyBase) or by downregulating raptor by RNAi (Fig. 2E,G; data not shown). Of note, TSC1/TSC2 overexpression seemed to reduce PSC cell size (Fig. 2E′). Conversely, TSC1 RNAi expression, which resulted in a larger PSC (Fig. 2F), did not significantly affect cell number but increased cell size (Fig. 2G,H). This suggests that TOR signaling not only supports PSC cell proliferation but also their growth. Finally we observed a strong drop in PSC cell number when we overexpressed Foxo (Fig. 2G), which is the main effector of IIS and whose targets are concomitantly regulated by the TOR kinase (Edgar, 2006). Together, these data indicate that IIS and TOR pathways are required in the PSC to promote niche cell proliferation/maintenance and growth.

Fig. 2.

IIS and TOR signaling cell-autonomously control the size of the PSC. (A-F′) Antp immunostaining (red) on lymph glands expressing the indicated transgene and a membrane-linked GFP (mCD8-GFP, green) under the control of col-Gal4. A′-F′ show high magnification views of the PSC region. Nuclei are stained with Topro3 (blue). Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bars: 100 μm. (G,H) Quantification of the number of cells per PSC (G) or the volume of the PSC (H) in control larvae and in larvae expressing the indicated transgenes in the PSC. Data are presented as box-and-whisker plots. Student’s t-test: ***P<0.0005; ****P<0.0001; NS, not significant.

Fig. 2.

IIS and TOR signaling cell-autonomously control the size of the PSC. (A-F′) Antp immunostaining (red) on lymph glands expressing the indicated transgene and a membrane-linked GFP (mCD8-GFP, green) under the control of col-Gal4. A′-F′ show high magnification views of the PSC region. Nuclei are stained with Topro3 (blue). Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bars: 100 μm. (G,H) Quantification of the number of cells per PSC (G) or the volume of the PSC (H) in control larvae and in larvae expressing the indicated transgenes in the PSC. Data are presented as box-and-whisker plots. Student’s t-test: ***P<0.0005; ****P<0.0001; NS, not significant.

Control of hematopoietic niche size by IIS and TOR signaling affects blood cell differentiation

Previous work showed that the PSC controls prohemocyte maintenance (Krzemien et al., 2007; Mandal et al., 2007). Notably, in contrast to what we observed in Pten larvae, increased PSC size has been associated with decreased differentiation (Mandal et al., 2007; Sinenko et al., 2009; Tokusumi et al., 2011). We thus assessed the effect of increased IIS/TOR signaling in the PSC on hemocyte differentiation. Although PI3Kcaax overexpression or TSC1 RNAi increased PSC size by more than twofold, P1 and Hnt immunostaining did not reveal an obvious reduction in either plasmatocyte or crystal cell differentiation (Fig. 3A-F). To ensure that we did not overlook a subtle phenotype, we measured crystal cell or plasmatocyte differentiation index on a minimum of 20 samples (see Materials and methods). We found that over-activation of the IIS or TOR pathway in the PSC significantly inhibited the differentiation of both type of hemocytes (Fig. 3G,H). Moreover, expression of an Hh-GFP reporter line (Tokusumi et al., 2011) was observed in all the PSC cells expressing either PI3Kcaax or TSC1 RNAi, suggesting that increased PSC size is associated with increased production of this inhibitor of prohemocyte differentiation (Mandal et al., 2007). Together, our results indicate that increased IIS/TOR signaling not only supports Drosophila hematopoietic niche growth but also promotes its activity.

Fig. 3.

Increasing IIS or TOR signaling in the PSC impairs blood cell differentiation. (A-F) Immunostaining against the plasmatocyte marker P1 (A-C) or the crystal cell marker Hnt (D-F) on lymph glands of third instar larvae expressing mCD8-GFP and the indicated transgene in the PSC (col-Gal4). Nuclei are stained with Topro3 (blue). Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bar: 100 μm. (G,H) Quantification of the crystal cell differentiation index (number of crystal cells per primary lobe/lobe volume) (G) and plasmatocyte differentiation index (Cg25C-GFP expression level per primary lobe/lobe volume) (H) in control larvae and in larvae expressing the indicated transgenes in the PSC. Arbitrary units. Student’s t-test: *P<0.05; **P<0.005; ***P<0.0005. (I-K) High magnification views of the PSC region showing Hh-GFP and Col expression.

Fig. 3.

Increasing IIS or TOR signaling in the PSC impairs blood cell differentiation. (A-F) Immunostaining against the plasmatocyte marker P1 (A-C) or the crystal cell marker Hnt (D-F) on lymph glands of third instar larvae expressing mCD8-GFP and the indicated transgene in the PSC (col-Gal4). Nuclei are stained with Topro3 (blue). Dashed red and yellow lines outline primary and secondary lobes, respectively. Scale bar: 100 μm. (G,H) Quantification of the crystal cell differentiation index (number of crystal cells per primary lobe/lobe volume) (G) and plasmatocyte differentiation index (Cg25C-GFP expression level per primary lobe/lobe volume) (H) in control larvae and in larvae expressing the indicated transgenes in the PSC. Arbitrary units. Student’s t-test: *P<0.05; **P<0.005; ***P<0.0005. (I-K) High magnification views of the PSC region showing Hh-GFP and Col expression.

IIS and TOR signaling cell-autonomously controls blood cell progenitor maintenance

Next, we modulated the activity of the IIS/TOR pathways specifically in the prohemocytes using the TepIV-Gal4 driver (Avet-Rochex et al., 2010). Reminiscent of the phenotypes observed in Pten larvae, over-activation of IIS by PI3Kcaax expression induced a massive differentiation of plasmatocytes (labeled with either Cg25C-GFP or P1) and crystal cells (labeled with either Hnt or Bc-GFP) in the primary and secondary lobes, paralleled by a strong reduction in the pool of prohemocytes (labeled with either Ance or a UAS-mCherry) (Fig. 4B,H; supplementary material Fig. S6B,G), and an overgrowth of the secondary lobes (supplementary material Fig. S5). Unexpectedly, knocking down IIS by expressing InR RNAi also favored prohemocyte differentiation (Fig. 4C,H; supplementary material Fig. S6C,G). However, in contrast to IIS activation, IIS knockdown reduced lymph gland growth (supplementary material Fig. S5) and did not cause lamellocyte differentiation (supplementary material Fig. S2). Finally, lowering TOR activity using raptor RNAi promoted hemocyte differentiation and significantly reduced the proportion of prohemocytes present in the primary lobes (Fig. 4D,H; supplementary material Fig. S6D,G). Similar phenotypes were observed when IIS and TOR signaling were reduced in the MZ using the dome-Gal4 driver (data not shown). Thus, a fine-tuning of IIS/TOR pathways in the blood cell progenitors seems to be crucial to prevent their differentiation and to control lymph gland growth. Furthermore, as increased PSC size in Pten zygotic mutants does not prevent the massive differentiation of the prohemocytes, we propose that over-activation of the IIS/TOR pathways in the MZ bypasses the progenitor-promoting activity of the PSC.

Fig. 4.

IIS and TOR signaling functions cell-autonomously in the prohemocytes to sense systemic diet restriction. (A-G,I-K) Third instar larval lymph glands. Plasmatocytes, crystal cells and prohemocytes are labeled with Cg25C-GFP, Hnt and Ance, respectively. Scale bar: 100 μm. (A-D′) TepIV-gal4 was used to drive the expression of the indicated transgene in the prohemocytes. (E-G,I-K) Diet restriction was induced either by starvation (F,J) or by depletion of slif activity in the fat body (ppl>slifanti) (G,K). (J′) Effect of starvation on Hnt and Ance expression in second larval instar lymph gland. (E-G) Nuclei are stained with Topro3. Dashed red and yellow lines outline primary and secondary lobes, respectively. (H) Proportion of prohemocytes (MZ area/primary lobe area) in the primary lobes of control larvae and in larvae expressing the indicated transgenes. Box-and-whisker representation. Student’s t-test: ***P<0.0005.

Fig. 4.

IIS and TOR signaling functions cell-autonomously in the prohemocytes to sense systemic diet restriction. (A-G,I-K) Third instar larval lymph glands. Plasmatocytes, crystal cells and prohemocytes are labeled with Cg25C-GFP, Hnt and Ance, respectively. Scale bar: 100 μm. (A-D′) TepIV-gal4 was used to drive the expression of the indicated transgene in the prohemocytes. (E-G,I-K) Diet restriction was induced either by starvation (F,J) or by depletion of slif activity in the fat body (ppl>slifanti) (G,K). (J′) Effect of starvation on Hnt and Ance expression in second larval instar lymph gland. (E-G) Nuclei are stained with Topro3. Dashed red and yellow lines outline primary and secondary lobes, respectively. (H) Proportion of prohemocytes (MZ area/primary lobe area) in the primary lobes of control larvae and in larvae expressing the indicated transgenes. Box-and-whisker representation. Student’s t-test: ***P<0.0005.

Starvation causes prohemocyte differentiation

One main function of the IIS pathway is to link tissue growth to nutrient sensing. Indeed, food shortage induces a reduction of insulin-like peptide (ILP) secretion by the insulin-producing cells (IPC) located in the brain (Edgar, 2006). The consecutive reduction in IIS signaling diminishes larval growth. The fat body acts as a nutrient sensor that restricts global growth through a humoral mechanism: upon diet restriction, ILP production by the IPC is reduced in response to the inactivation of the TOR pathway in the fat body. Accordingly, depletion of the amino acid transporter Slimfast (Slif) in the fat body disrupts TOR signaling and is sufficient to mimic diet restriction (Colombani et al., 2003). As IIS/TOR signaling is active in the lymph gland and controls prohemocyte maintenance, we investigated whether the larval nutritional status alters lymph gland homeostasis. When the larvae were starved for 24 hours, we observed a strong increase in plasmatocyte differentiation as revealed by Cg25C-GFP expression, and a concomitant decrease in Ance expression, indicative of a switch from prohemocyte maintenance towards differentiation (Fig. 4F,J). Unexpectedly, only a few crystal cells were present in the lymph glands of starved third instar larvae and we observed that they burst (Fig. 4J; data not shown). However, the examination of starved second instar larvae revealed the precocious differentiation of crystal cells associated with a decreased in the MZ (Fig. 4J′). Hence, starvation induces precocious differentiation of prohemocytes into both crystal cells and plasmatocytes. Of note, it is possible that starvation also directly affects differentiated hemocytes, notably crystal cells. We then tested whether food shortage was sensed directly by the prohemocytes or mediated by a systemic humoral response. To do so, we mimicked amino-acid deprivation by inhibiting Slif expression either in the prohemocytes or in the fat body, where its downregulation causes a systemic reduction in ILP levels (Colombani et al., 2003). Whereas expression of the RNA antisense slifAnti in the prohemocytes did not affect their maintenance (supplementary material Fig. S6E,G), its expression with the fat body-specific pumpless-Gal4 (ppl-Gal4) driver caused massive plasmatocyte differentiation and decreased the prohemocyte pool (Fig. 4G,H,K; supplementary material Fig. S6F). Therefore, the systemic response to nutrient availability controls blood cell progenitor maintenance in the lymph gland.

Conclusions

Our results demonstrate that IIS/TOR signaling plays a dual role in the maintenance of the blood cell progenitors by acting both within the hematopoietic niche to control its size and its activity, and within the prohemocytes to control their fate. To gain a comprehensive view of IIS/TOR function in Drosophila hematopoiesis and in light of the recent report showing that differentiated hemocytes can feedback on prohemocyte maintenance (Mondal et al., 2011), it will be interesting to explore the role of these pathways in the differentiated blood cells. In addition, our data are consistent with a model whereby the IIS/TOR pathways link prohemocyte maintenance to the Drosophila larvae nutritional status. We speculate that food shortage, by sensitizing blood cell progenitors to differentiation, might affect the cellular immune response. Along this line, the rate of encapsulation of parasitoid wasp eggs, which relies primarily on the differentiation of lamellocytes, has been shown to diminish in larvae that were deprived of yeast before infestation (Vass and Nappi, 1998). We anticipate that future studies will allow further understanding of how developmental and environmental cues are integrated by IIS/TOR signaling to control blood cell homeostasis.

We thank H. Hafen, P. Léopold, J. Colombani, L. Pick, N. Tapon, M. Crozatier, I. Ando and Bloomington and Vienna Drosophila stock centers for flies and reagents. We thank B. Ronsin for developing the Volocity macro. We thank Toulouse RIO imaging platform and B. Augé for assistance with confocal, and members of our team for discussions.

Funding

This work was supported by the Centre National de la Recherche Scientifique (CNRS) and by grants from the Agence Nationale pour la Recherche and the Association pour la Recherche sur le Cancer [1114]. B.B. was supported by a CNRS-PED fellowship.

Amcheslavsky
A.
,
Jiang
J.
,
Ip
Y. T.
(
2009
).
Tissue damage-induced intestinal stem cell division in Drosophila
.
Cell Stem Cell
4
,
49
61
.
Avet-Rochex
A.
,
Boyer
K.
,
Polesello
C.
,
Gobert
V.
,
Osman
D.
,
Roch
F.
,
Auge
B.
,
Zanet
J.
,
Haenlin
M.
,
Waltzer
L.
(
2010
).
An in vivo RNA interference screen identifies gene networks controlling Drosophila melanogaster blood cell homeostasis
.
BMC Dev. Biol.
10
,
65
.
Chell
J. M.
,
Brand
A. H.
(
2010
).
Nutrition-responsive glia control exit of neural stem cells from quiescence
.
Cell
143
,
1161
1173
.
Colombani
J.
,
Raisin
S.
,
Pantalacci
S.
,
Radimerski
T.
,
Montagne
J.
,
Léopold
P.
(
2003
).
A nutrient sensor mechanism controls Drosophila growth
.
Cell
114
,
739
749
.
Crozatier
M.
,
Ubeda
J. M.
,
Vincent
A.
,
Meister
M.
(
2004
).
Cellular immune response to parasitization in Drosophila requires the EBF orthologue collier
.
PLoS Biol.
2
,
E196
.
Edgar
B. A.
(
2006
).
How flies get their size: genetics meets physiology
.
Nat. Rev. Genet.
7
,
907
916
.
Hartenstein
V.
(
2006
).
Blood cells and blood cell development in the animal kingdom
.
Annu. Rev. Cell Dev. Biol.
22
,
677
712
.
Huang
H.
,
Potter
C. J.
,
Tao
W.
,
Li
D. M.
,
Brogiolo
W.
,
Hafen
E.
,
Sun
H.
,
Xu
T.
(
1999
).
PTEN affects cell size, cell proliferation and apoptosis during Drosophila eye development
.
Development
126
,
5365
5372
.
Ikeya
T.
,
Galic
M.
,
Belawat
P.
,
Nairz
K.
,
Hafen
E.
(
2002
).
Nutrient-dependent expression of insulin-like peptides from neuroendocrine cells in the CNS contributes to growth regulation in Drosophila
.
Curr. Biol.
12
,
1293
1300
.
Jung
S. H.
,
Evans
C. J.
,
Uemura
C.
,
Banerjee
U.
(
2005
).
The Drosophila lymph gland as a developmental model of hematopoiesis
.
Development
132
,
2521
2533
.
Krzemien
J.
,
Dubois
L.
,
Makki
R.
,
Meister
M.
,
Vincent
A.
,
Crozatier
M.
(
2007
).
Control of blood cell homeostasis in Drosophila larvae by the posterior signalling centre
.
Nature
446
,
325
328
.
Krzemien
J.
,
Crozatier
M.
,
Vincent
A.
(
2010a
).
Ontogeny of the Drosophila larval hematopoietic organ, hemocyte homeostasis and the dedicated cellular immune response to parasitism
.
Int. J. Dev. Biol.
54
,
1117
1125
.
Krzemien
J.
,
Oyallon
J.
,
Crozatier
M.
,
Vincent
A.
(
2010b
).
Hematopoietic progenitors and hemocyte lineages in the Drosophila lymph gland
.
Dev. Biol.
346
,
310
319
.
Lanot
R.
,
Zachary
D.
,
Holder
F.
,
Meister
M.
(
2001
).
Postembryonic hematopoiesis in Drosophila
.
Dev. Biol.
230
,
243
257
.
Leevers
S. J.
,
Weinkove
D.
,
MacDougall
L. K.
,
Hafen
E.
,
Waterfield
M. D.
(
1996
).
The Drosophila phosphoinositide 3-kinase Dp110 promotes cell growth
.
EMBO J.
15
,
6584
6594
.
Mandal
L.
,
Martinez-Agosto
J. A.
,
Evans
C. J.
,
Hartenstein
V.
,
Banerjee
U.
(
2007
).
A Hedgehog- and Antennapedia-dependent niche maintains Drosophila haematopoietic precursors
.
Nature
446
,
320
324
.
McLeod
C. J.
,
Wang
L.
,
Wong
C.
,
Jones
D. L.
(
2010
).
Stem cell dynamics in response to nutrient availability
.
Curr. Biol.
20
,
2100
2105
.
Meister
M.
,
Lagueux
M.
(
2003
).
Drosophila blood cells
.
Cell Microbiol.
5
,
573
580
.
Minakhina
S.
,
Steward
R.
(
2010
).
Hematopoietic stem cells in Drosophila
.
Development
137
,
27
31
.
Mondal
B. C.
,
Mukherjee
T.
,
Mandal
L.
,
Evans
C. J.
,
Sinenko
S. A.
,
Martinez-Agosto
J. A.
,
Banerjee
U.
(
2011
).
Interaction between differentiating cell- and niche-derived signals in hematopoietic progenitor maintenance
.
Cell
147
,
1589
1600
.
Oldham
S.
,
Stocker
H.
,
Laffargue
M.
,
Wittwer
F.
,
Wymann
M.
,
Hafen
E.
(
2002
).
The Drosophila insulin/IGF receptor controls growth and size by modulating PtdInsP(3) levels
.
Development
129
,
4103
4109
.
Owusu-Ansah
E.
,
Banerjee
U.
(
2009
).
Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation
.
Nature
461
,
537
541
.
Puig
O.
,
Marr
M. T.
,
Ruhf
M. L.
,
Tjian
R.
(
2003
).
Control of cell number by Drosophila FOXO: downstream and feedback regulation of the insulin receptor pathway
.
Genes Dev.
17
,
2006
2020
.
Sinenko
S. A.
,
Mandal
L.
,
Martinez-Agosto
J. A.
,
Banerjee
U.
(
2009
).
Dual role of wingless signaling in stem-like hematopoietic precursor maintenance in Drosophila
.
Dev. Cell
16
,
756
763
.
Sorrentino
R. P.
,
Tokusumi
T.
,
Schulz
R. A.
(
2007
).
The Friend of GATA protein U-shaped functions as a hematopoietic tumor suppressor in Drosophila
.
Dev. Biol.
311
,
311
323
.
Sousa-Nunes
R.
,
Yee
L. L.
,
Gould
A. P.
(
2011
).
Fat cells reactivate quiescent neuroblasts via TOR and glial insulin relays in Drosophila
.
Nature
471
,
508
512
.
Tapon
N.
,
Ito
N.
,
Dickson
B. J.
,
Treisman
J. E.
,
Hariharan
I. K.
(
2001
).
The Drosophila tuberous sclerosis complex gene homologs restrict cell growth and cell proliferation
.
Cell
105
,
345
355
.
Tennessen
J. M.
,
Thummel
C. S.
(
2011
).
Coordinating growth and maturation-insights from Drosophila
.
Curr. Biol.
21
,
R750
R757
.
Tokusumi
T.
,
Tokusumi
Y.
,
Hopkins
D. W.
,
Shoue
D. A.
,
Corona
L.
,
Schulz
R. A.
(
2011
).
Germ line differentiation factor Bag of Marbles is a regulator of hematopoietic progenitor maintenance during Drosophila hematopoiesis
.
Development
138
,
3879
3884
.
Vass
E.
,
Nappi
A. J.
(
1998
).
The effects of dietary yeast on the cellular immune response of Drosophila melanogaster against the larval parasitoid, Leptopilina boulardi
.
J. Parasitol.
84
,
870
872
.
Vilmos
P.
,
Nagy
I.
,
Kurucz
E.
,
Hultmark
D.
,
Gateff
E.
,
Andó
I.
(
2004
).
A rapid rosetting method for separation of hemocyte sub-populations of Drosophila melanogaster
.
Dev. Comp. Immunol.
28
,
555
563
.
Yilmaz
O. H.
,
Valdez
R.
,
Theisen
B. K.
,
Guo
W.
,
Ferguson
D. O.
,
Wu
H.
,
Morrison
S. J.
(
2006
).
Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells
.
Nature
441
,
475
482
.
Zhang
J.
,
Grindley
J. C.
,
Yin
T.
,
Jayasinghe
S.
,
He
X. C.
,
Ross
J. T.
,
Haug
J. S.
,
Rupp
D.
,
Porter-Westpfahl
K. S.
,
Wiedemann
L. M.
, et al. 
. (
2006
).
PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention
.
Nature
441
,
518
522
.

Competing interests statement

The authors declare no competing financial interests.

Supplementary information