You are on page 1of 8

Blackwell Science, LtdOxford, UKCMICellular Microbiology 1462-5822Blackwell Publishing Ltd, 200359573580Review ArticleDrosophila blood cellsM. Meister and M.

Lagueux

Cellular Microbiology (2003) 5(9), 573–580 doi:10.1046/j.1462-5822.2003.00302.x

Microreview

Drosophila blood cells

Marie Meister and Marie Lagueux* mechanisms. In this context, Drosophila has been the
UPR 9022 du CNRS, IBMC, 15 rue René Descartes, model of choice in the two last decades, to unravel the
67084 Strasbourg, France. molecular mechanisms of non-adaptive host defence.
Drosophila immunity includes two complementary facets
which are humoral and cellular immunity. Most of the
Summary
recent progress relates to Drosophila humoral immunity.
Drosophila blood cells or haemocytes belong to three Infection by bacteria or fungi triggers the rapid and mas-
lineages: plasmatocytes, crystal cells and lamello- sive production in the fat body, which is the liver counter-
cytes. There is no equivalent of a lymphoid lineage in part in insects, of a number of antimicrobial peptides and
insects which have no adaptive immunity. Haemato- other effectors. Two major signalling pathways control this
poiesis is under the control of a number of transcrip- process which are called the Toll and the Imd pathways
tion factors and signalling pathways (such as GATA (reviewed in Hoffmann and Reichhart, 2002). The intrac-
factors, JAK/STAT or Notch pathways) most of which ellular components of both pathways, as well as the extra-
have homologues which participate in the control of cellular cascades that lead to their activation, are currently
mammalian haematopoiesis. Drosophila plasmato- under intense investigation in various laboratories.
cytes are professional phagocytes reminiscent of the Opposed to this, cellular immunity, the second facet of
cells from the mammalian monocyte/macrophage lin- Drosophila host defence, has only recently attracted
eage. Several receptors responsible for recognition of renewed interest. We would like in this review to give an
microorganisms or apoptotic corpses have been update of our current knowledge on haematopoiesis in
identified, which include a Scavenger Receptor, a Drosophila, and on the functions of the various blood cell
CD36 homologue and a peptidoglycan recognition or haemocyte types.
protein. Crystal cells contain the enzymes necessary
for humoral melanization that accompanies a number
I. Haematopoiesis in Drosophila
of immune reactions. The production of melanin gen-
erates, as by-products, cytotoxic free radicals that are Two phases of haematopoiesis
believed to participate in the killing of pathogens.
Drosophila haematopoiesis occurs in two phases during
Finally, lamellocytes represent a cell type that specif-
development, and gives rise to three haemocyte lineages
ically differentiates after parasitism of Drosophila
that share characteristics with mammalian myeloid lin-
larvae and forms a capsule around the invader.
eages. Drosophila, like all arthropods, has no equivalent
Encapsulation together with melanization eventually
of a lymphoid lineage. A first haematopoietic wave takes
kill the parasite within the capsule.
place in the second half of embryogenesis when a popu-
lation of haemocytes originate in the procephalic meso-
Introduction derm and migrate to colonize the whole embryo along
invariant paths (Tepass et al., 1994). These cells, called
Innate immunity is phylogenetically the oldest defence
plasmatocytes, act as macrophages as they eliminate
system in the animal kingdom. It evolved before adaptive
apoptotic cells (Franc et al., 1996; 1999). A second pop-
immunity which has arisen only recently in evolution and
ulation differentiates simultaneously nearby the anterior
is the hallmark of vertebrate immunity. Like all inverte-
region of the gut where it remains localized around the
brates, insects defend themselves through innate immune
proventriculus (Lebestky et al., 2000). These cells are
crystal cells (see below) and their role in the embryo is
unknown. Towards the end of embryogenesis, the precur-
Received 31 March, 2003; revised 2 May, 2003; accepted 7 May,
2003. *For correspondence. E-mail M.Meister@ibmc.u-strasbg.fr; sors of the lymph glands form in the lateral mesoderm,
Tel. (+33) 03 88 41 70 32; Fax (+33) 03 88 60 69 22. and migrate dorsally to prefigure the first paired lobes of

© 2003 Blackwell Publishing Ltd


574 M. Meister and M. Lagueux
the organ (Rugendorff et al., 1994). The lymph glands are lation: they probably correspond to a variation on the
the main site of haematopoiesis during larval stages plasmatocyte theme as intermediate forms are often
(Shrestha and Gateff, 1982; Rizki and Rizki, 1984; Lanot found in the lymph glands. Finally, a third independent
et al., 2001). They are composed of a variable number of haemocyte lineage exists in larvae, but is seldom
paired lobes that are located along the dorsal vessel observed in healthy animals. These cells called lamello-
(Fig. 1). In the posterior lobes they contain essentially cytes differentiate massively in the lymph glands after
undifferentiated precursor cells called prohaemocytes. parasitization and are large flat cells devoted to encapsu-
More anteriorly they mostly contain fully differentiated lation of invaders too large to be phagocytosed by plas-
haemocytes which are released into the circulation. Cir- matocytes (Fig. 1). A commonly encountered immune
culating haemocytes (Fig. 1) comprise a majority of plas- threat for Diptera (flies) is parasitism by Hymenoptera
matocytes which are the dedicated phagocytes, and a (wasps) species that lay their eggs in Diptera larvae (Car-
small proportion (<5%) of crystal cells. Crystal cells con- ton et al., 1986). The cellular reaction that fends off para-
tain crystalline inclusions that correspond to enzymes sites thus involves the production of a specific, inducible
necessary for humoral melanization (see below). An addi- haemocyte lineage.
tional cell type is found in the anteriormost lobes of the At the onset of metamorphosis, the lymph glands
lymph glands that displays features of intense protein release large numbers of active phagocytes that are
synthesis. These ‘secretory cells’ are never seen in circu- called pupal macrophages and play a crucial role in tissue

Fig. 1. Drosophila haemocytes.


A. Scanning electron microscopy of a larval
haematopoietic organ. The lymph glands are
organized as paired lobes along the dorsal ves-
sel (arrow). Anterior is to the left.
B. Circulating haemocytes stained with DAPI.
Plasmatocytes are small round cells whereas
lamellocytes (arrows) are larger and flattened.
C. Phase-contrast microscopy of circulating
crystal cells (arrows) with conspicuous crystal-
line inclusions.
D. Confocal image of phagocytosis of E. coli
(K12 Alexa Fluor 594) by GFP-labelled plasma-
tocytes from a tep1-GFP larva.
E–G. Encapsulation of parasitoid wasp eggs/
larvae. A wasp larva is surrounded by lamello-
cytes (blue nuclei in E); the cell layer becomes
thicker while melanization of the parasite is ini-
tiated (arrowhead in F); eventually the wasp
egg/larva is totally melanized within the lamel-
locyte capsule (G). Lamellocytes are visualized
with a lacZ enhancer trap marker in E and F.
H–J. Transmission electron microscopy of a
plasmatocyte (H), a crystal cell (I) and lamello-
cytes (J). Bars: 50 mm (A, E–G), 20 mm (B),
10 mm (C, D), 2 mm (H–J).

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


Drosophila blood cells 575
remodelling as they phagocyte cells of doomed larval at an early embryonic stage, and later for gene expression
structures. In this process they also eat up the remainder during their maturation. Several transcription factors and
of the lymph glands, and at later stages no haematopoi- signalling pathways have been demonstrated to govern
etic organ can be found (Lanot et al., 2001). This modifi- lineage specification in Drosophila haematopoiesis. Two
cation of the properties of plasmatocytes and the Zinc-finger transactivators, Gcm (Glial Cells Missing, Ber-
simultaneous dispersal of the lymph glands is under the nardoni et al., 1997; Lebestky et al., 2000) and Gcm2
control of the steroid hormone ecdysone (Lanot et al., (Alfonso and Jones, 2002) are required for plasmatocyte
2001; Sorrentino et al., 2002) which orchestrates all tissu- fate in the embryo, and this has also been documented
lar modifications related to metamorphosis. In Drosophila for Gcm at the larval stage. The crystal cell fate is
adults, the only haemocyte type that is present is the instructed in larvae by the Serrate/Notch pathway (Duvic
plasmatocyte, and the pool of adult haemocytes likely et al., 2002; Lebestky et al., 2003) and the Runx1-related
derives from larval plasmatocytes. Given their morpholog- Lozenge transcrition factor (Lebestky et al., 2000). The
ical and functional features, it is generally considered that Friend-of-GATA homologue U-shaped antagonises crystal
Drosophila plasmatocytes resemble the mammalian cell development (Fossett et al., 2001). Transcription fac-
monocyte/macrophage lineage. tors that determine lamellocyte development are not yet
identified. Mutations in three genes have long been known
to stimulate lamellocyte production: these are Toll and
Transcriptional control of haematopoiesis
JAK dominant gain-of-function, and cactus loss-of-
Our understanding of regulation of haemocyte prolifera- function mutations (Lemaitre et al., 1995; Luo et al., 1995;
tion and specification comes from genetic analysis of Qiu et al., 1998). It is however, possible that the effect of
mutants (Fig. 2). Control of haemocyte proliferation was these mutations on haemocyte differentiation is indirect.
investigated at the larval stage and it was recently shown It is striking that most of the players in Drosophila hae-
that the unique Drosophila homologue of PDGFR/ matopoiesis identified so far are counterparts of gene
VEGFR, named PVR, together with one of its three puta- products that are also central to mammalian haemato-
tive ligands PVF2, plays a crucial role in the control of poiesis, such as GATA factors, the Notch pathway, Runx1
prohaemocyte proliferation (Munier et al., 2002). This find- or the JAK/STAT pathway to cite but a few examples. It
ing was essentially demonstrated by the dramatic effect appears that the same building blocks are used with dif-
of PVF2 overexpression in larvae, which resulted in a 300- ferent combinations to control haematopoiesis in inverte-
fold increase in blood cell counts, due to excessive prolif- brates and vertebrates.
eration of prohaemocytes at the expense of differentiation.
A comparable (40-fold) effect was obtained by pan-hae-
II. Haemocyte functions
matopoietic overexpression of an activated form of Ras,
and this effect is mediated by the Raf/MAPK pathway Drosophila that are deprived of haemocytes are clearly
(Asha et al., 2003). It is however, not documented yet sensitized to infection. This was shown in larvae carrying
whether the proliferation signal produced by PVF2/PVR is a mutation called domino which results in extremely
transmitted via the Ras/Rak/MAPK pathway. This prolifer- reduced haemocyte counts. When this mutation is com-
ation effect of PVF2/PVR observed at larval stages does bined with mutations affecting humoral immunity (imd) or
not apply to the embryonic stage where PVF/PVR were melanization, resistance of larvae to infection is strongly
rather proposed to establish a guidance system respon- decreased compared to single mutants (Braun et al.,
sible for the migration of the plasmatocytes throughout the 1998). Similarly, inactivation of adult haemocytes by mas-
embryo (Cho et al., 2002). sive injection of polystyrene beads reduced the ability of
Early data have also implicated the JAK/STAT and the imd flies to fight bacterial infection (Elrod-Erickson et al.,
Toll pathways in the control of circulating haemocyte num- 2000). Haemocytes are responsible for a number of
bers as gain-of-function mutations of JAK and of Toll result immune functions in Drosophila, among which phagocy-
in increased blood cell counts (Luo et al., 1995; Lemaitre tosis, encapsulation and melanization have been docu-
et al., 1995), whereas loss-of-function mutations in the Toll mented. We have outlined the current data on these
pathway have the opposite effect (Qiu et al., 1998). immune reactions below.
Haemocyte identity in embryos is specified by the GATA
factor Serpent (Srp; Rehorn et al., 1996; Waltzer et al.,
Phagocytosis
2002). A similar function for srp is likely to occur at larval
stages as its expression is recorded before all differentia- In vertebrates, the elimination of microorganisms and the
tion markers in prohaemocytes (Lebestky et al., 2000). It removal of apoptotic cells are achieved by the same
is proposed that srp might be required both for the spec- phagocytes. However, whereas phagocytosis of non-self
ification of haemocyte primordium within the mesoderm particles induces inflammation, that of apoptotic cells

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


576 M. Meister and M. Lagueux
either downregulates or suppresses inflammation (Vandi- A scavenger receptor, dSR-CI with similar broad ligand
vier et al., 2002). Phagocytosis occurs in several steps. recognition as mammalian class A Scavenger Receptor
The first consists in the attachment of the phagocyte to was identified by Pearson et al. (1995). dSR-CI can medi-
the targeted particle followed by alteration of the cytosk- ate binding to both Gram-negative and Gram-positive bac-
eleton and internalization. The engulfed target is then teria when transfected into CHO cells and participates to
destroyed within phagosomes by lysosomal enzymes, some extent to the binding of these microorganisms to S2
reactive oxygen species and nitric oxide (Jones et al., cells, a primary Drosophila blood cell culture (Rämet
1999). The latter phases are likely conserved between et al., 2001). PGRP-LC, a Peptidoglycan Recognition Pro-
invertebrates and vertebrates, involving namely actin- tein, mediates phagocytosis of Gram-negative, but not
remodelling and vesicule trafficking (Rämet et al., 2002a). Gram-positive bacteria by S2 cells (Rämet et al., 2002a).
It has been proposed that phagocytosis of both apop- Interestingly, PGRP-LC belongs to a family of 12 members
totic cells and microorganisms requires two processes: in Drosophila (Werner et al., 2000). This putative mem-
tethering, followed by actin-dependent engulfment brane bound protein is involved in the induction of the fat
(reviews in Aderem and Underhill, 1999; Fadok and body Imd transduction pathway, leading to the production
Chimini, 2001; Hoffmann et al., 2001; Greenberg and of antibacterial peptides after an infection with Gram-neg-
Grinstein, 2002). Tethering of the target to the phagocyte ative bacteria (Gottar et al., 2002). Similarly, PGRP-SA
is achieved by receptors that will either directly recognize which is a soluble PGRP, is required for the proper induc-
determinants on microorganisms or apoptotic cells, or tion of the Toll pathway after infection with Gram-positive
bind through opsonizing molecules. Most of these recep- bacteria (Michel et al., 2001). Gottar et al. (2002) have
tors have a broad recognition spectrum for both endoge- suggested that PGRPs are sensors which define the type
nous and exogenous molecules, and may participate in of microbial infection and drive the proper signalling path-
the phagocytosis of both types of targets. Mammalian way. An additional family of recognition proteins found in
examples are the Scavenger Receptor family (qualified as insects are the Gram-negative bacteria-binding proteins
‘molecular fly papers’ by Krieger et al., 1993). Tethering or GNBPs which all contain gluconase-like domains.
favours a tight association between the target and the Three cDNAs encoding GNBPs have been cloned from a
phagocyte, and clustering of the binding proteins then Drosophila blood cell line library. Although their role in
mediates phagocytosis. The release of the signal trigger- phagocytosis has not been demonstrated, it was shown
ing the appropriate on/off switch of inflammation seems that one GNBP (DGNBP-1) can bind LPS and b-1,3-
to be dependent on the engulfment receptor that is simul- glucan (Kim et al., 2000). Although there is good evidence
taneously recruited. A few receptors are able to initiate that Croquemort, dSC-RI or PGRP-LC can mediate
engulfment, and they also participate in the recognition of phagocytosis of target particles, there is no demonstration
the target. For ingestion of microorganisms, well studied that they can by themselves trigger internalization without
receptors are the Mannose Receptor, that binds sugar assistance from co-receptors.
moieties on microorganisms, two proteins of the integrin The Drosophila genome harbours six genes encoding
family, CR3 (aMb2) and CR4 (aXb2) that are receptors thiolester-containing proteins or TEPs (Lagueux et al.,
for the opsonizing complement factor C3bi, and the Fcg 2000). These proteins are related to the a2macroglobulin/
receptors that recognize antibodies. Phagocytosis of apo- complement factor C3 family as they share the 12 signa-
ptotic cells is dependent on the recognition of phosphati- ture domains characteristic of these proteins, which com-
dylserine by its receptor (PS-R), but can also be mediated prise the well conserved thiolester motif region. However,
by two integrins aVb3 and aVb5, the tyrosine kinase MER TEPs clearly form a new group in this family due to a
and CD91 (Henson et al., 2001; Huynh et al., 2002). distinct cystein arrangement in their C-terminal region. All
In Drosophila, plasmatocytes are responsible for the putative TEPs possess a signal peptide indicating that
disposal of both microorganisms (Fig. 1) and apoptotic they are secreted proteins. The transcription of three out
cells. The mechanisms by which they recognize and of six tep genes (tep1, 2 and 4) is upregulated after an
engulf their targets are poorly understood. Phagocytosis immune challenge and tep1 and tep4 are mainly
of apoptotic cells at the embryonic stage requires wild- expressed in haemocytes (Lagueux et al., 2000; and M.
type function of the croquemort gene, which encodes a Lagueux, unpubl. obs.). We have proposed that TEPs
CD36 homologue (Franc et al., 1996, 1999). In mammals function during an immune response either as opsonins
CD36 is a class B Scavenger Receptor which acts in to promote phagocytosis, in a C3-like manner, or as pro-
concert with the vitronectin receptor (aVb3) and PS-R to tease inhibitors, in an a2-macroglobulin manner. The best
engulf apoptotic corpses (Fadok et al., 1998). A homo- evidence so far, in favour of the opsonin hypothesis,
logue of PS-R has also been described in Drosophila but comes from studies in an other insect, Anopheles gam-
no functional studies have yet demonstrated a role for this biae. Levashina et al. (2001) found similar TEP proteins
receptor in apoptotic cell removal (Henson et al., 2001). in this model insect and could show that phagocytosis of

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


Drosophila blood cells 577
Gram-negative bacteria in an Anopheles blood cell line is ate into an adapted haemocyte lineage. The identity of the
strongly reduced when transcription of the atep1 gene is signal has yet to be determined. Thus lamellocytes, to
impaired. some extent, exhibit adaptive characteristics as they only
It is likely that plasmatocytes also send signals to other differentiate in response to a specific immune challenge.
immunocompetent tissues. When larvae are infected per They form several layers of cells around the parasite, to
os with the phytopathogenic Erwinia carotovora, they which they are attracted either by the previously attached
respond by the activation in the fat body of the Imd path- plasmatocytes, or by wasp determinants. The nature of
way with subsequent production of antibacterial peptides this mechanism is also unknown.
(Basset et al., 2000). In mutant larvae with a reduced
number of haemocytes, such as domino larvae for
Melanization
instance, the Imd pathway is not activated in response to
infection. This indicates that haemocytes act as messen- Humoral melanization in arthropods produces black pig-
gers that somehow convey information to the fat body in ment as a result of the activation of a biochemical pathway
the case of bacterial infection. However, the nature of the that converts tyrosine to melanin (reviews in Ashida and
signal is still unknown. Brey, 1997; Söderhäll and Cerenius, 1998). Melanization
is controlled by a cascade of serine proteases that ulti-
mately cleave the zymogen prophenoloxidase to its active
Encapsulation
form. Phenoloxidase then catalyses the oxidation of phe-
Some 50 hymenopteran species are reported parasites of nols to quinones, which polymerize non-enzymatically to
Drosophila (Carton et al., 1986). The wasp females lay form melanin. It was shown in lepidopterans that the cas-
their eggs in the haemocoel of young larvae. This foreign cade is activated by initial recognition of non-self molec-
body is detected by plasmatocytes which are the first line ular patterns such as b-1,3-glucans, peptidoglycan or
immune supervisors in circulation. They readily attach to LPS. The different elements of the cascade are not yet
the chorion of the egg (Russo et al., 1996) and a few elucidated in Drosophila, but a key control serpin has
hours later a strong cellular reaction is observed in the been recently identified that restricts phenoloxidase activ-
haematopoietic organ with enhanced proliferation, ity to the site of injury or infection (De Gregorio et al.,
increase in crystal cell numbers (Sorrentino et al., 2002), 2002; Ligoxygakis et al., 2002). Serpin-27 A regulates the
and massive differentiation of lamellocytes (Lanot et al., melanization cascade through the specific inhibition of
2001). Lamellocytes then form a multilayered capsule prophenoloxidase processing by the terminal serine pro-
around the invader, which is ultimately accompanied by tease. Three genes encoding prophenoloxidases [Fujim-
blackening due to melanization (Fig. 1). Within the cap- oto et al., 1995; Berkeley Drosophila Genome Project
sule, the parasite is eventually killed, by asphyxiation or (BDGP)] are present in the Drosophila genome, and in
by the local production of cytotoxic free radicals, quinones larvae they are specifically expressed in crystal cells (M.
or semiquinones (Nappi et al., 1995; 2000). The whole Meister, unpubl. obs.), which, once activated, readily dis-
process of parasite encapsulation raises several intriguing rupt and deliver their content into the haemolymph where
questions. The first relates to the signal that is produced the enzymes can function. A number of intermediate com-
by plasmatocytes once they have recognized an invader pounds formed during melanin synthesis are cytotoxic
which they cannot phagocyte. This signal is perceived by (see above, killing of the parasite), it is thus important to
prohaemocytes in the lymph glands which then differenti- strictly control the localization of the reaction. This is

Fig. 2. Regulation of Drosophila haematopoie-


sis. Blood cell identity is conferred by the GATA
factor Serpent (srp). Proliferation is controlled
by the PVF2/PVR, the Ras/Raf, the Toll and the
JAK/STAT pathways. Crystal cell specification
is under the control of the Notch (N) pathway
and the Runx1 homologue Lozenge (lz) trans-
activator, and antagonized by the Friend-of-
GATA homologue U-shaped (ush). Plasmato-
cytes are specified by two Glial-cells-missing
(gcm) transactivators, then further differentiate
into pupal macrophages under the impulse of
the steroid hormone ecdysone (Ecd). Lamello-
cyte specification requires wild-type functions
of Notch and of the JAK/STAT pathway.

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


578 M. Meister and M. Lagueux
achieved not only by the regulatory serpins, but also by the increasing numbers of blood cell markers. It also
the fact that activated phenoloxidase shows a tendency should help to gain insight, in the future, into molecular
to aggregate (Ashida and Brey, 1997). mechanisms of functions that are the hallmarks of mye-
Melanization was proposed to participate in the sealing loid-type blood cells.
of a wound before the more elaborate epithelial wound
healing process (Lai-Fook, 1966) that takes several days.
This is based namely on the observation that Drosophila Acknowledgements
melanization-deficient mutants show defects in wound The authors thank Jules Hoffmann for continued support. Work
healing with excessive bleeding and reduced survival in their laboratory is supported by CNRS, NIH grant 1PO1
(Rämet et al., 2002b). These observations could indicate AI44220-02 to A. Ezekowitz and J. Hoffmann, the French Min-
a role for melanization in the coagulation process, or that istère de l’Education Nationale, de la Recherche et de la Tech-
the proteolytic cascades leading to melanization and to nologie, EntoMed, Exelixis and l’Association de la Recherche
contre le Cancer.
coagulation share components. Nothing is known to date
on molecular mechanisms responsible for coagulation in
Drosophila. It will thus be challenging to determine how References
melanization and coagulation are activated in this model,
and how closely they are related. Aderem, A., and Underhill, D.M. (1999) Mechanisms of
phagocytosis in macrophages. Annu Rev Immunol 17:
593–623.
Extracellular matrix production Alfonso, T.B., and Jones, B.W. (2002) gcm2 promotes glial
cell differentiation and is required with glial cells missing
Drosophila haemocytes are known to produce abundant for macrophage development in Drosophila. Devel Biol
extracellular matrix material. Indeed many proteins have 248: 369–383.
Asha, H., Nagy, I., Kovacs, G., Stetson, D., Ando, I., and
been purified from the supernatant of Kc cell cultures
Dearolf, C.R. (2003) Analysis of ras-induced overprolifera-
which are of haematopoietic origin (review in Fessler tion in Drosophila hemocytes. Genetics 163: 203–215.
et al., 1994; Nelsson et al., 1994; Goto et al., 2001). Ashida, M., and Brey, P. (1997) Recent advances in research
Among these, laminins, collagen IV, tenascinm, glutactin, on the insect propheno-loxidase cascade. In Molecular
peroxidasin and haemolectin were subsequently found Mechanisms of Immune Response in Insects. Brey, P.T.,
and Hultmark, D. (eds). London: Chapman & Hall, pp. 135–
to be strongly expressed in embryonic and larval
172.
haemocytes. However, the almost complete absence of Basset, A., Khush, R., Braun, A., Gardan, L., Boccard, F.,
haemocytes in domino larvae does not result in apparent Hoffmann, J.A., and Lemaitre, B. (2000) The phytopatho-
structural tissue defects and so the significance of this genic bacteria Erwinia carotovora infects Drosophila and
synthesis is not yet clear. Some of these molecules exhibit activates an immune response. Proc Natl Acad Sci USA
97: 3376–3381.
peculiar features, namely peroxidasin which contains a
Bernardoni, R., Vivancos, V., and Giangrande, A. (1997)
putative peroxidase domain associated with several leu- glide/gcm is expressed and required in the scavenger cell
cine-rich repeats similar to those found in the extracellular lineage. Development 191: 118–130.
domain of Toll family members (Nelsson et al., 1994). Braun, A., Hoffmann, J.A., and Meister, M. (1998) Analysis
Haemolectin displays similarity with the von Willebrand of the Drosophila host defense in domino mutant larvae,
which are devoid of hemocytes. Proc Natl Acad Sci USA
factor which in vertebrates plays a role in haemostasis
95: 14337–14342.
(Goto et al., 2001). However, as no mutants for both these Carton, Y., Bouletreau, M., Van Lenteren, J.C., and van
genes are available, their function has not been assessed Alphen, J.C.M. (1986) The Drosophila parasitoid wasps. In
to date. The Genetics and Biology of Drosophila. Ashburner, M.,
Carson, H.L., and Thompson, J.N. (eds). New York: Aca-
demic Press, pp. 347–394.
Cho, N.K., Keyes, L., Johnson, E., Heller, J., Ryner, L.,
Conclusion Karim, F., and Krasnow, M.A. (2002) Developmental con-
The field of Drosophila immunity has witnessed significant trol of blood cells migration by the Drosophila VEGF path-
way. Cell 108: 865–876.
development over the last few years, thereby fuelling our De Gregorio, E., Han, S.J., Lee, W.J., Baek, M.J., Osaki, T.,
general understanding of innate immunity. Opposed to Kawabata, S., et al. (2002) An immune-responsive Serpin
this, for a long time Drosophila has failed to be a leading regulates the melanization cascade in Drosophila. Dev Cell
model in the analysis of haematopoiesis and of blood cell 3: 581–592.
functions. A number of recent studies have now demon- Duvic, B., Hoffmann, J.A., Meister, M., and Royet, J. (2002)
Notch signaling controls lineage specification during
strated that it has a role to play in the understanding of Drosophila larval hematopoiesis. Current Biol 12: 1923–
proliferation, commitment and differentiation of haemato- 1927.
poietic precursors, thanks to its powerful genetics and to Elrod-Erickson, M., Mishra, S., and Schneider, D. (2000)

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


Drosophila blood cells 579
Interactions between the cellular and humoral immune M., and Resnick, D. (1993) Molecular flypaper, host
responses in Drosophila. Current Biol 10: 781–784. defense, and atherosclerosis. Structure, binding proper-
Fadok, V.A., Bratton, D.L., Frasch, S.C., Warner, W.L., and ties, and functions of macrophage scavenger receptors. J
Henson, P.M. (1998) The role of phosphatidylserine in Biol Chem 268: 4569–4572.
recognition of apoptotic cells by phagocytes. Cell Death Lagueux, M., Perrodou, E., Levashina, E.A., Capovilla, M.,
Differ 5: 551–562. and Hoffmann, J.A. (2000) Constitutive expression of a
Fadok, V.A., and Chimini, G. (2001) The phagocytosis of complement-like protein in Toll and JAK gain-of-function
apoptotic cells. Semin Immunol 13: 365–372. mutants of Drosophila. Proc Natl Acad Sci USA 97: 11427–
Fessler, L.I., Nelsson, R.E., and Fessler, J.H. (1994) 11432.
Drosophila extracellular matrix. Methods Enzymol 245: Lai-Fook, J. (1966) The repair of wounds in the integument
271–294. of insects. J Insect Physiol 12: 195–226.
Fossett, N., Tevosian, S.G., Gajewski, K., Zhang, Q., Orkin, Lanot, R., Zachary, D., Holder, F., and Meister, M. (2001)
S.H., and Schulz, R.A. (2001) The friend of GATA proteins Post-embryonic hematopoiesis in Drosophila. Dev Biol
U-shaped, FOG-1, and FOG-2 function as negative regu- 230: 243–257.
lators of blood, heart, and eye development in Drosophila. Lebestky, T., Chang, T., Hartenstein, V., and Banerjee, U.
Proc Natl Acad Sci USA 98: 7342–7347. (2000) Specification of Drosophila hematopoietic lineage
Franc, N.C., Dimarcq, J.L., Lagueux, M., Hoffmann, J.A., and by conserved transcription factors. Science 288: 146–149.
Ezekowitz, R.A.B. (1996) Croquemort, a novel Drosophila Lebestky, T., Jung, S.H., and Banerjee, U. (2003) A Serrate-
hemocyte/macrophage receptor that recognizes apoptotic expressing signaling center controls Drosophila hemato-
cells. Immunity 4: 431–443. poiesis. Genes Dev 17: 348–353.
Franc, N.C., Heitzler, P., Ezekowitz, R.A.B., and White, K. Lemaitre, B., Meister, M., Govind, S., Georgel, P., Steward,
(1999) Requirement for croquemort in phagocytosis of R., Reichhart, J.M., and Hoffmann, J.A. (1995) Functional
apoptotic cells in Drosophila. Science 284: 1991–1994. analysis and regulation of nuclear import of dorsal during
Fujimoto, K., Okino, N., Kawabata, S., Iwanaga, S., and the immune response in Drosophila. EMBO J 14: 536–545.
Ohnishi, E. (1995) Nucleotide sequence of the cDNA Levashina, E.A., Moita, L.F., Blandin, S., Vriend, G.,
encoding the proenzyme of phenol oxidase A1 of Droso- Lagueux, M., and Kafatos, F.C. (2001) Conserved role of
phila melanogaster. Proc Natl Acad Sci USA 92: 7769– a complement-like protein in phagocytosis revealed by
7773. dsRNA knockout in cultured cells of the mosquito, Anoph-
Goto, A., Kumagai, T., Kumagai, C., Hirose, J., Narita, H., eles gambiae. Cell 104: 709–718.
Mori, H., et al. (2001) A Drosophila haemocyte-specific Ligoxygakis, P., Pelte, N., Ji, C., Leclerc, V., Duvic, B., Belvin,
protein, hemolectin, similar to human von Willebrand factor. M., et al. (2002) A serpin mutant links Toll activation to
Biochem J 359: 99–108. melanization in the host defence of Drosophila. EMBO J
Gottar, M., Gobert, V., Michel, T., Belvin, M., Duyk, G., Hoff- 21: 6330–6337.
mann, J.A., Ferrandon, D., and Royet, J. (2002) The Droso- Luo, H., Hanratty, W.P., and Dearolf, C.R. (1995) An amino
phila immune response against Gram-negative bacteria is acid substitution in the Drosophila hopTum–l Jak kinase
mediated by a peptidoglycan recognition protein. Nature causes leukemia-like hematopoietic defects. EMBO J 14:
416: 640–644. 1412–1420.
Greenberg, S., and Grinstein, S. (2002) Phagocytosis and Michel, T., Reichhart, J.M., Hoffmann, J.A., and Royet, J.
innate immunity. Curr Opin Immunol 14: 136–145. (2001) Drosophila Toll is activated by Gram-positive bacte-
Henson, P.M., Bratton, D.L., and Fadok, V.A. (2001) Apop- ria through a circulating peptidoglycan recognition protein.
totic cell removal. Curr Biol 11: R795–R805. Nature 414: 756–759.
Hoffmann, P.R., deCathelineau, A.M., Ogden, C.A., Lever- Munier, A.I., Doucet, D., Perrodou, E., Zachary, D., Meister,
rier, Y., Bratton, D.L., Daleke, D.L., et al. (2001) Phos- M., Hoffmann, J.A., Janeway, C.A. Jr and Lagueux, M.
phatidylserine (PS) induces PS receptor-mediated (2002) PVF2, a PDGF/VEGF-like growth factor, induces
macropinocytosis and promotes clearance of apoptotic hemocyte proliferation in Drosophila larvae. EMBO Rep 3:
cells. J Cell Biol 155: 649–659. 1195–1200.
Hoffmann, J.A., and Reichhart, J.M. (2002) Drosophila innate Nappi, A.J., Vass, E., Frey, F., and Carton, Y. (1995)
immunity: an evolutionary perspective. Nat Immunol 3: Superoxide anion generation in Drosophila during melan-
121–126. otic encapsulation of parasites. Eur J Cell Biol 68: 450–
Huynh, M.L., Fadok, V.A., and Henson, P.M. (2002) 456.
Phosphatidylserine-dependent ingestion of apoptotic cells Nappi, A.J., Vass, E., Frey, F., and Carton, Y. (2000) Nitric
promotes TGF-beta1 secretion and the resolution of oxide involvement in Drosophila immunity. Nitric Oxide 4:
inflammation. J Clin Invest 109: 41–50. 423–430.
Jones, S.L., Lindberg, F.P., and Brown, E.J. (1999) Phago- Nelsson, R.E., Fessler, L.I., Takagi, Y., Blumberg, B., Keene,
cytosis. In Fundamental Immunology, 4th edn. Paul, W.E. D.R., Olson, P.F., Parker, C.G., and Fessler, J.H. (1994)
(ed). Philadelphia: Lippincott-Raven Publishers, pp. 997– Peroxidasin: a novel enzyme-matrix protein of Drosophila
1020. development. EMBO J 13: 3438–3447.
Kim, Y.S., Ryu, J.H., Han, S.J., Choi, K.H., Nam, K.B., Jang, Pearson, A., Lux, A., and Krieger, M. (1995) Expression
I.H., et al. (2000) Gram-negative bacteria-binding protein, cloning of dSR-CI, a class C macrophage-specific scaven-
a pattern recognition receptor for lipopolysaccharide and ger receptor from Drosophila melanogaster. Proc Natl
beta-1,3-glucan that mediates the signaling for the induc- Acad Sci USA 92: 4056–4060.
tion of innate immune genes in Drosophila melanogaster Qiu, P., Pan, P.C., and Govind, S. (1998) A role for the
cells. J Biol Chem 275: 32721–32727. Drosophila Toll/cactus pathway in larval hematopoiesis.
Krieger, M., Acton, S., Ashkenas, J., Pearson, A., Penman, Development 125: 1909–1520.

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580


580 M. Meister and M. Lagueux
Rämet, M., Manfruelli, P., Pearson, A., Mathey-Prevot, B., Shrestha, R., and Gateff, E. (1982) Ultrastructure and
and Ezekowitz, R.A. (2002a) Functional genomic analysis cytochemistry of the cell types in the larval hematopoietic
of phagocytosis and identification of a Drosophila receptor organs and hemolymph of Drosophila melanogaster. Dev
for E. coli. Nature 416: 644–648. Growth Differ 24: 65–82.
Rämet, M., Lanot, R., Zachary, D., and Manfruelli, P. (2002b) Söderhäll, K., and Cerenius, L. (1998) Role of the prophe-
JNK signaling pathway is required for efficient wound heal- noloxidase-activating system in invertebrate immunity. Curr
ing in Drosophila. Dev Biol 241: 145–156. Opin Immunol 10: 23–28.
Rämet, M., Pearson, A., Manfruelli, P., Li, X., Koziel, H., Sorrentino, R.P., Carton, Y., and Govind, S. (2002) Cellular
Gobel, V., Chung, E., Krieger, M., and Ezekowitz, R.A. immune response to parasite infection in the Drosophila
(2001) Drosophila scavenger receptor CI is a pattern lymph gland is developmentally regulated. Dev Biol 243:
recognition receptor for bacteria. Immunity 15: 1027– 65–80.
1038. Tepass, U., Fessler, L.I., Aziz, A., and Hartenstein, V.
Rehorn, K.P., Thelen, H., Michelson, A.M., and Reuter, R. (1994) Embryonic origin of hemocytes and their relation-
(1996) A molecular aspect of hematopoiesis and endo- ship to cell death in Drosophila. Development 120:
derm development common to vertebrates and Drosophila. 1829–1837.
Development 122: 4023–4031. Vandivier, R.W., Ogden, C.A., Fadok, V.A., Hoffmann, P.R.,
Rizki, T.M., and Rizki, R.M. (1984) The cellular defense sys- Brown, K.K., Botto, M., et al. (2002) Role of surfactant
tem of Drosophila melanogaster. In Insect Ultrastructure, proteins A, D, and C1q in the clearance of apoptotic cells
Vol. 2. King, RC, and Akai, H. (eds). Plenum Publishing in vivo and in vitro: calreticulin and CD91 as a common
Corporation, pp. 579–604. collectin receptor complex. J Immunol 169: 3978–3986.
Rugendorff, A., Younossi-Hartenstein, A., and Hartenstein, Waltzer, L., Bataille, L., Peyrefitte, S., and Haenlin, M. (2002)
V. (1994) Embryonic origin and differentiation of the Droso- Two isoforms of Serpent containing either one or two GATA
phila heart. Roux’s Arch Dev Biol 203: 266–280. zinc fingers have different roles in Drosophila haemato-
Russo, J., Dupas, S., Frey, F., Carton, Y., and Brehelin, M. poiesis. EMBO J 21: 5477–5486.
(1996) Insect immunity: early events in the encapsulation Werner, T., Liu, G., Kang, D., Ekengren, S., Steiner, H., and
process of parasitoid (Leptopilina boulardi) eggs in resis- Hultmark, D. (2000) A family of peptidoglycan recognition
tant and susceptible strains of Drosophila. Parasitology proteins in the fruit fly Drosophila melanogaster. Proc Natl
112: 135–142. Acad Sci USA 97: 13772–13777.

© 2003 Blackwell Publishing Ltd, Cellular Microbiology, 5, 573–580

You might also like