the lesswright mutation activates rel-related proteins, leading to overproduction of larval...

14

Click here to load reader

Upload: liang-huang

Post on 30-Oct-2016

215 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

www.elsevier.com/locate/ydbio

Developmental Biology

The lesswright mutation activates Rel-related proteins, leading to

overproduction of larval hemocytes in Drosophila melanogaster

Liang Huanga, Shunji Ohsakob, Soichi Tandaa,TaDepartment of Biological Sciences and Molecular and Cell Biology Program, Ohio University, Athens, OH 45701, USA

bDepartment of Basic Techniques and Facilities, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu, Tokyo 183-8526, Japan

Received for publication 20 September 2004, revised 4 February 2005, accepted 5 February 2005

Abstract

The lesswright (lwr) gene encodes an enzyme that conjugates a small ubiquitin-related modifier (SUMO). Since the conjugation of

SUMO occurs in many different proteins, a variety of cellular processes probably require lwr function. Here, we demonstrate that lwr

function regulates the production of blood cells (hemocytes) in Drosophila larvae. lwr mutant larvae develop many melanotic tumors in the

hemolymph at the third instar stage. The formation of melanotic tumors is due to a large number of circulating hemocytes, which is

approximately 10 times higher than those of wild type. This overproduction of hemocytes is attributed to the loss of lwr function primarily in

hemocytes and the lymph glands, a hematopoietic organ in Drosophila larvae. High incidences of Dorsal (Dl) protein in the nucleus were

observed in lwr mutant hemocytes, and the dl and Dorsal-related immunity factor (Dif) mutations were found to be suppressors of the lwr

mutation. Therefore, the lwr mutation leads to the activation of these Rel-related proteins, key transcription factors in hematopoiesis. We also

demonstrate that dl and Dif play different roles in hematopoiesis. dl primarily stimulates plasmatocyte production, but Dif controls both

plasmatocyte and lamellocyte production.

D 2005 Elsevier Inc. All rights reserved.

Keywords: Drosophila hematopoiesis; dorsal; Dorsal-related immunity factor; lesswright; SUMO conjugation; plasmatocyte; lamellocyte

Introduction

The innate immune system is evolutionarily ancient

and common among most eukaryotes and consists of

humoral and cellular components (Evans et al., 2003;

Hoffmann et al., 1999; Hultmark, 1993; Kimbrell and

Beutler, 2001). In Drosophila, the humoral response

primarily represents the production of antimicrobial

peptides in the fat body. The cellular innate immunity is

handled by circulating blood cells capable of recognizing

and neutralizing foreign objects. These cells also work as

scavengers of apoptotic cells in normal development.

Because the balance of these functions is so important,

the proliferation and differentiation of hemocytes must be

tightly regulated.

0012-1606/$ - see front matter D 2005 Elsevier Inc. All rights reserved.

doi:10.1016/j.ydbio.2005.02.006

T Corresponding author. Fax: +1 740 593 0300.

E-mail address: [email protected] (S. Tanda).

The Drosophila larval hemocyte population consists of

several cell types, some of which reside only in a blood–cell-

forming organ, the lymph gland (Fig. 1A; Lanot et al., 2001;

Rizki, 1978). Three cell types, plasmatocytes, crystal cells,

and lamellocytes, are present in hemolymph. The majority

are plasmatocytes, which are able to phagocytose microbes

and apoptotic cells (Fig. 1B). A small fraction (b5%) are

crystal cells, which are involved in melanization reactions.

Rarely found in circulation in normal circumstances,

lamellocytes are key defensive players when larvae are

infested by parasitoid wasp eggs (Fig. 1C; Lanot et al., 2001;

Rizki and Rizki, 1984; Sorrentino et al., 2002). In addition to

these cells, prohemocytes (hematopoietic stem cells) and

secretory cells are found in the lymph glands (Lanot et al.,

2001). A small number of prohemocytes circulate in

hemolymph. The population structure of hemocytes changes

as larvae develop and undergo metamorphosis (Lanot et al.,

2001; Rizki, 1957). Several genes have been found to direct

280 (2005) 407–420

Page 2: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

Fig. 1. Lymph gland, plasmatocyte, and lamellocyte in Drosophila larva.

(A) The right side of the lymph gland of the third instar larva. Lamellocytes

are visible with msn-lacZ marker (blue cells). An arrow indicates the

position of the dorsal vessel, and the cells marked with asterisks are

pericardial cells that associate with the dorsal vessel. A large arrowhead

indicates the first lobe of the lymph gland and small arrowheads point the

secondary lobes. (B) Plasmatocytes in circulation aremsn-lacZ negative. (C)

Large and matured lamellocyte is msn-lacZ positive (blue pigments in the

nucleus). Images were taken with DIC optics, and bars correspond to 20 Am.

L. Huang et al. / Developmental Biology 280 (2005) 407–420408

differentiation of specific hemocytes. A Drosophila GATA

homologue, serpent, is required for the expression of two

lineage-specific genes, glial cell missing (gcm) and lozenge

(lz) (Lebestky et al., 2000). The gcm gene encodes a

transcription factor and is required for the plasmatocyte

lineage. A Drosophila acute myeloid leukemia-1 homo-

logue, lz, and the Notch pathway are required for the crystal

cell lineage (Duvic et al., 2002; Lebestky et al., 2003).

Recently, yantar and collier were found to direct lamellocyte

differentiation (Crozatier et al., 2004; Sinenko et al., 2004).

However, precise mechanisms of the proliferation and

differentiation of hemocytes still remain elusive.

Three signal transduction pathways are known to influ-

ence the number of hemocytes in circulation. They are the

Ras, JAK/STAT, and Tl pathways (Asha et al., 2003; Harrison

et al., 1995; Luo et al., 1997; Qiu et al., 1998). Mutations that

activate these pathways increase the total hemocyte numbers

10- to 100-fold and often stimulate lamellocyte production in

the absence of parasitoid wasp infestation. Ras functions in

theMAP kinase pathway and stimulates cell division. It is one

of the most common oncogenes found in many different

human cancers (Bos, 1989; McCormick, 1994). When the

Drosophila ras1 gene is overexpressed in the lymph gland

and hemocytes, total hemocyte counts increase nearly 100-

fold (Asha et al., 2003). The JAK-STAT pathway is a

conserved signal transduction pathway and plays a variety of

roles in Drosophila and many other eukaryotes including

humans (Kisseleva et al., 2002; O’Shea et al., 2002; Ward et

al., 2000). A dominant mutation of JAK, hopscotchTum-l,

causes a drastic increase in plasmatocytes as well as

lamellocytes in a temperature-dependent manner. Similarly,

dominant mutations of Tl, Tl10B, and Tl3, produce a large

number of hemocytes (Braun et al., 1997; Qiu et al., 1998). In

this case, the lamellocyte population increases to 10–20% of

the entire hemocyte population. Several genes in the Tl

signaling pathway also exhibit a similar defect when mutated.

Although it is not clear if these signal transduction pathways

possess lineage specific functions, they play some general

role in Drosophila hematopoiesis.

The studies of the NF-nB pathway, discovered in 1986

(Sen and Baltimore, 1986), have made a huge contribution to

the understanding of mammalian immune systems, partic-

ularly in acquired (or adaptive) immunity (Baldwin, 1996;

Ghosh et al., 1998; Hatada et al., 2000). NF-nB is a dimer of

members of the Rel-related transcription factors. A unique

aspect in the NF-nB pathway is that Inhibitors of nB, InBs,keep the transcription factor NF-nB in the cytoplasm from its

action in the nucleus. Ubiquitination, thus degradation, of

InB is required for the activation of the NF-nB pathway. A

Drosophila Rel-related gene, dl, was discovered to be an

important component in the establishment of the embryonic

dorsal–ventral axis (Nusslein-Volhard et al., 1980; Steward,

1987) and was found to be a part of Tl signaling (Anderson et

al., 1985; Schupbach, 1987). The activation of Tl signaling

starts from the cell surface receptor Tl and ends at the

entrance of the Dl transcription factor into the nucleus. The

function of Tl signaling in innate immunity was later

discovered indirectly. Sequence analysis of several antimi-

crobial peptide genes spotted nB sites, binding sites for NF-

nB and Dl, in their 5Vregulatory regions (Engstrom et al.,

1993), which connected the Tl pathway to humoral

immunity. Discovery and subsequent studies of other Rel-

related genes, Dif and Relish, further support a key role of

the Tl pathway in Drosophila immunity (Dushay et al.,

1996; Ip et al., 1993). Furthermore, the homology between

mammalian InBs and Cactus (Cact) (Geisler et al., 1992;

Kidd, 1992) demonstrates that the parallelism extends to the

regulatory mechanism of mammalian and Drosophila NF-

nB activity. The presence of multiple Rel-related proteins

and Tl-like receptors in Drosophila as well as in other

eukaryotes makes the system more complex, but versatile in

innate immune responses and hematopoiesis. In the last

decade, remarkable progress has been made toward the

understanding of Drosophila humoral immunity including

the role of the Tl pathway (Hoffmann and Reichhart, 2002;

Meister et al., 1997; Silverman and Maniatis, 2001; Tzou et

al., 2002). On the other hand, the regulatory mechanisms of

cellular immunity are much less understood.

Recently, ubiquitin’s small cousins, SUMO molecules,

were discovered to add another layer of complexity in NF-

nB signaling (Desterro et al., 1998). SUMO conjugation

(sumoylation) regulates a variety of cellular functions

(Melchior, 2000; Muller et al., 2001; Yeh et al., 2000).

Proteins subjected to sumoylation are involved in onco-

Page 3: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420 409

genesis, transcriptional regulation, nuclear transport, and

others. Ubiquitin and SUMO molecules are biochemically

quite similar to each other. Furthermore, they require a set of

similar modifying enzymes, activating (E1), conjugating

(E2), and ligating (E3) enzymes, which show high con-

servation among most eukaryotes. The similarity between

ubiquitination and sumoylation creates an interesting sit-

uation where the same lysine residues on a given protein can

be modified by both ubiquitin and SUMO. Thus, ubiquiti-

nation and sumoylation can counteract each other. This

scenario is seen in the regulation of InBa degradation

(Desterro et al., 1998). In addition to NF-nB signaling, the

activity of the JAK/STAT pathway might be regulated in part

by sumoylation since a Protein inhibitor of activated STAT

(Pias) possesses a RING finger motif, an E3 SUMO ligase

signature (Schmidt and Muller, 2003; Shuai and Liu, 2003).

However, these possibilities have not yet been explored in

Drosophila hematopoiesis.

Here, we report that the Lwr protein, a SUMO conjugase,

plays an important role in regulation of larval hematopoiesis

in Drosophila melanogaster. Recessive as well as dominant

negative mutations of the lwr gene result in the over-

proliferation of hemocytes in larvae. We found that the loss

of lwr function led to the accumulation of the Rel-related

protein Dl in the nuclei of circulating hemocytes, and that dl

and Dif mutations are suppressors of the lwr mutation.

Taken together, our results indicate that the lwr function is

inhibitory to dl and Dif activities. Furthermore, we

demonstrate that dl and Dif possess different properties in

hematopoiesis, particularly in lamellocyte production.

Materials and methods

Drosophila culture conditions and stocks

Flies were cultured in JAZZ mix (Fisher Scientific)

supplemented with inactive brewer’s yeast (SAP Product

Corporation) and soy flour (ADM). JAZZ mix was cooked

in a steam kettle according to the manufacturer’s instruc-

tions. The stocks were maintained at room temperature, and

the experiments were conducted in uncrowded conditions at

258C except for the experiment with the e33CGAL4 driver,

which was carried out at 288C.Hypomorphic alleles of lwr, lwr4-3, and lwr5, were

introduced to lethal free backgrounds of P{neoFRT}40A

and b1 cn1 bw1, respectively, in order to minimize effects

of unwanted hidden mutations linked to these lwr alleles

(Sun et al., 2003). The deficiency Df(2L)J4 is associated

with a small, cytologically-invisible deletion at 36C8-9

(Meng et al., 1999), and the deficiency Df(2L)TW119

deletes a small section between 36C4-2 and 36E1. These

deficiencies remove both the dl and Dif genes and were

used to create the dl Dif double mutant combination. The

CgGAL4, the e33CGAL4, and the Lsp2GAL4 drivers were

described previously (Asha et al., 2003; Cherbas et al.,

2003; Harrison et al., 1995). The UAS-Dif and the UAS-

Tl10B lines were generous gifts from Y.T. Ip, and the UAS-

Tl10B lines were described elsewhere (Hu et al., 2004).

Other mutations and aberrations used in this study are

described in FlyBase (http://flybase.bio.indiana.edu/).

Transgenics

pUAS transgene constructs (see below) were amplified

on a large scale and purified using MAXI Prep kit (Qiagen).

DNA was resuspended in doubly distilled water at a

concentration of approximately 500 Ag/ml. We used the y

w; Sb, P{D2-3}99B/ TM6 stock as host (Robertson et al.,

1988). Germline transformation was performed as described

(Spradling, 1986; Sullivan et al., 2000).

UAS-dl construct: A full-length dorsal cDNA in the

pBluescript KS (Rushlow et al., 1989) was excised with the

restriction enzymes KpnI and XbaI. This fragment was then

cloned into the pUAST vector (Brand and Perrimon, 1993)

between the KpnI and XbaI sites.

UAS-lwrDN construct: We constructed a dominant

negative form of lwr by replacing cys93 and leu97 in the

conserved SUMO conjugation site with arginine and alanine

residues, respectively. This strategy was successfully

applied to the mouse Ubc9 gene (Tashiro et al., 1997). A

single-stranded template for in vitro mutagenesis (Sculptor,

Amersham) was obtained from lwr cloned in the pBluscript

SK vector (STRATAGENE). The primer sequence for

mutagenesis was the following: 5V-CGGGCACCGTTCG-CCTGTCGCTGGCCGACGAGG. The introduced muta-

tions were confirmed by DNA sequencing. The lwrDN

allele was then isolated by PCR with primers with added

BamHI and XbaI sites and cloned into the pUAST vector

between the BglII and XbaI sites. The forward primer with

BamHI site and the reverse primer with XbaI site were the

following: 5V-CGGGATCCACCATGTCCGGCATTGCTand 5V-GCTCTAGATTTTATTGAAATTACATAGGTT. We

tested a UAS-lwrDN transgene on the third chromosome for

its ability to counteract the wild type function. We crossed it

with the e22cGAL4 and the tubulinGAL4 drivers. In both

cases, about 18% of embryos, excluding unfertilized eggs,

did not hatch of the 1000–1500 embryos examined. These

numbers were substantially higher than those with aUAS-lwr

(wild type) transgene (b3%). Furthermore, some exhibited

cuticle defects similar to those of lwr mutants (Epps and

Tanda, 1998). Therefore, we conclude that this lwrDN allele

can be used to mimic effects of the lwr mutation.

Genotyping larvae

Genotypes of larvae homozygous for a given second-

linked mutation were determined using the CyO balancer

with a yellow+ ( y+) transgene in a y background. The

mutant larvae were distinguished by a lesser degree of

pigmentation of the cephalopharyngeal skeleton than

those of heterozygous siblings. A similar approach was

Page 4: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420410

used on the third-linked mutations using the TM6B

balancer with Tubby (Tb), or the TM3 balancer with an

actin-GFP transgene. The presence of GPF in larvae

was monitored using a Nikon SMZ1000 stereoscopic

microscope equipped with an epi-fluorescent apparatus.

Hemocyte counting

Egg collection was done daily, and the larvae were raised

to the mid/late third instar stage. Feeding larvae with still-

retracted anterior spiracles were harvested and used for this

study. They are presumed to be those before receiving the

first pulse of ecdysone, which triggers wandering

behavior and stimulates hemocyte production. We found

that variations in total blood cell counts were much larger

among wandering larvae than feeding larvae.

Hemocytes were counted using a hemacytometer, and the

total hemocyte counts were presented as the number of

hemocytes per milliliter of hemolymph. Larvae were rinsed

well in water and blotted on Kimwipes to remove excess

water before bleeding. A small incision was made near the

posterior spiracles and the hemolymph was directly loaded

on a hemacytometer. After placing a coverslip over the

hemolymph, all hemocytes but crystal cells were counted

using differential-interference-contrast (DIC) optics at a

magnification of 200�.

Statistical tests

Averages of hemocyte counts were compared by t test or

analysis of variance (ANOVA). In most cases, we evaluated

the differences between experimental genotype and corre-

sponding internal control by t test. In order to assess the

effects on total hemocyte counts among different genotypes,

we applied ANOVA to our data sets. First, we compared the

averages of the controls by ANOVA and confirmed that

there were no differences among the controls. When the

differences among different genotypes were significant,

Bonferroni’s multiple comparison test was used to compare

the averages of different genotypes.

Histological procedures

Paraffin sections were prepared using standard procedure

(Presnell and Schreibman, 1997). Late third instar larvae

were rinsed well in water and fixed in FAAG (80% EtOH,

4% Formaldehyde, 5% Glacial acetic acid, 1% Glutaralde-

hyde) for 15 min at room temperature. Small incisions were

made 2 min after the larvae were immersed in the fixing

solution. The specimens were then transferred to a

scintillation vial containing fresh FAAG and further fixed

overnight at 48C. Fixed larvae were then processed

according to standard procedure for paraffin sections.

Sectioning (5 Am thick) was done with a regular microtome

equipped with a disposable razor blade. Mayer Hematox-

ylin/Eosin Y staining was done using standard procedure.

Dehydrated specimens were mounted with Permount (Fisher

Scientific).

h-galactosidase staining was done according to the

method described in Sullivan et al. (2000). Larvae were

bled as described above and hemolymph was smeared on a

22 � 22 mm coverslip. Hemocytes were briefly dried and

then fixed for 20 min with 4% formaldehyde in Phosphate

Buffered Saline (PBS). The specimens were rinsed a few

times in PBS (5 min each) and then stained for

h-galactosidase activity overnight at 378C. After staining,

hemocytes were rinsed in PBS and mounted in 70%

glycerol.

For a Giemsa-stained blood smear, the hemolymph was

bled directly into a drop of 2 Al of PBS on a glass slide,

spread using a pair of forceps and dried. The blood smear

was fixed in 100%methanol for 5 min and stained for 20 min

in 10% Giemsa stain (Sigma) in water. The specimens were

rinsed in water for a few minutes and destained in 2 � 10�4

N HCl for 75 s. After being rinsed in water, the blood smear

was air-dried and mounted in Permount (Fisher Scientific).

Immunohistochemistry on hemocytes was carried out on

the cells smeared on a coverslip. The cells were first dried

for 20 min and fixed in 3.7% formaldehyde/PBS at room

temperature. The specimens were washed for 3 min four

times in PBS. The cells were then permeabilized in 0.1%

Triton X-100/PBS for 5 min and washed for 3 min three

times in PBS. After permeabilization, the cells were

incubated in 5% normal goat serum/PBS (blocking solution)

for 30 min at room temperature. Antibodies were diluted in

the blocking solution, and hemocytes were incubated with

primary antibodies overnight at 48C in a moist chamber. The

specimens were washed for 10 min five times at room

temperature in PBS and then incubated for 1 h at room

temperature with secondary antibodies diluted in the block-

ing solution. The cells were washed for 10 min 5 times at

room temperature and mounted with VectaShield (Vector

Laboratory) or Prolong (Molecular Probes). Anti-Dl mono-

clonal antibodies (concentrated form from Developmental

Studies Hybridoma Bank) were diluted 100-fold. Anti-

phospho-Histone H3 antibodies (1 Ag/Al, Upstate) were

diluted 200-fold. Secondary antibodies conjugated with

either Alexa Fluor 488 or 594 (Molecular Probes) were

diluted either 500- or 1000-fold.

A Nikon Optiphot-2 equipped with an epi-fluorescent

apparatus was used for all specimens. Images were captured

using a XM1200 digital camera (Nikon) and assembled

using Adobe PhotoShop.

Results

lesswright mutants develop melanotic tumors due to

overproduction of hemocytes

Mutants with amorphic lwr alleles die around the early

third instar larval stage (Sun et al., 2003). Mutant larvae

Page 5: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

Fig. 2. Melanotic tumors observed in lwr mutants. Panel A shows typical melanotic masses observed in lwr mutant larvae near the end of their larval lives.

Panel B shows an aggregate of overproliferated hemocytes in the hemocoel. Many lamellocytes (indicated with an arrow), which are flat in shape, surround the

mass although the cell types in the center of the mass cannot be unambiguously determined. The fat body (indicated with arrowheads) are also visible in the

same panel. Panel C shows hemocytes invading the fat body (indicated with arrowheads). Melanization associated with hemocytes (indicated with an arrow) is

seen in the central portion of the fat body, suggesting the presence of crystal cells that supply phenoloxidase and its substrate. The bars in panels B and C

correspond to 25 Am.

L. Huang et al. / Developmental Biology 280 (2005) 407–420 411

with hypomorphic alleles develop to the late third instar

stage but rarely pupate, surviving an additional few days as

larvae. They develop melanotic tumors during this third

instar stage. The tumors are usually free in the hemolymph

and become very large in the posterior half of the body (Fig.

2). In some cases, we observed excess hemocytes invade

self-tissues such as fat body cells (Fig. 2). Such invasion is

probably attributed to the abnormal property of hemocytes

due to the loss of lwr function. It can alternatively be

interpreted as resulting from abnormal properties of the fat

body due to loss of lwr function since abnormal basement

membranes can be targeted by normal hemocytes (Rizki and

Rizki, 1974).

To investigate why lwr mutant larvae develop melanotic

tumors, we first measured the number of circulating

hemocytes of lwr mutants as well as several controls

including Oregon-R and Tl10B (Table 1). Total hemocyte

counts of wild type and heterozygous controls varied from

2.1� 106 to 4.1� 106 per ml of hemolymph. The number of

Table 1

Hematopoietic defects of lwr, Tl10B, dl, and Dif mutants and their double and tri

Genotype Controla

Total hemocyte FSDb (�106/ml

of hemolymph)

Oregon-R 2.1 F 1.08 (35)

Canton-S 4.1 F 1.54 (35)

Tl10B/+ N/Ac

lwr4-3/lwr5 2.2 F 1.12 (35)

lwr4-3 dl1/lwr5 Df(2L)J4 2.0 F 0.61 (35)

lwr4-3 Dif2/lwr5 Df(2L)J4 2.1 F 0.69 (35)

lwr5 Df(2L)J4/lwr4-3 Df(2L)TW119 2.2 F 0.89 (35)

dl1/Df(2L)J4 2.0 F 0.99 (35)

Dif2/Df(2L)J4 2.2 F 0.87 (35)

Df(2L)J4/Df(2L)TW119 2.1 F 0.82 (35)

Note. The averages of total hemocyte counts excluding crystal cells were pres

deficiencies Df(2L)J4 and Df(2L)TW119 delete both the dl and Dif genes.a Heterozygous larvae of mixed genotypes in culture.b Standard deviation.c Not applicable.d Statistically significant differences between mutant and control ( P b 0.001).e Statistically insignificant differences between mutant and control.f Not determined.

hemocytes of lwr mutant larvae was 23.0 � 106 per ml of

hemolymph, which was statistically different from that of the

heterozygous control animals (2.2 � 106 per ml of

hemolymph, P b 0.001 in t test). This high hemocyte count

was very similar to that of Tl10B mutants (20.1�106 per ml of

hemolymph). Therefore, as in Tl10B and Tl3 (Qiu et al., 1998),

an excess number of circulating hemocytes probably caused

the development of melanotic tumors in lwr mutant larvae.

Since three different hemocytes are known to be in the

hemolymph, we also examined how hemocyte populations

were affected in these mutant larvae. We omitted crystal

cells from this study because they usually burst within 1 min

or less after bleeding, which leads to inaccurate estimates of

crystal cells. We estimated the proportion of lamellocytes in

the total circulating hemocytes as a parameter to represent

differences in the hemocyte population. Lamellocytes were

distinguished by using a lamellocyte specific marker, msn-

lacZ (Fig. 1C; Braun et al., 1997), or by their characteristic

morphology (Rizki, 1957). The percentage of lamellocytes

ple combinations

Mutant

Total hemocyte FSDb (�106/ml

of hemolymph)

% Lamellocyte FSDb

N/Ac 0.7 F 0.46 (15)

N/Ac 1.0 F 1.26 (15)

20.1 F 5.76 (32) 7.9 F 4.04 (11)

23.0 F 3.00 (35)d 27.5 F 5.24 (15)

10.0 F 2.79 (35)d 44.1 F 3.69 (15)

6.8 F 2.18 (35)d 12.2 F 6.87 (15)

1.9 F 1.34 (35)e 2.1 F 1.46 (15)

2.0 F 0.82 (35)e N/Df

2.4 F 1.43 (35)e N/Df

1.8 F 0.96 (35)e N/Df

ented. The number of larvae examined is indicated in parentheses. The

Page 6: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

Table 2

Effect of a dominant negative lwr gene on the number of hemocytes

GAL4 driver Controla Experimental

Total hemocytes FSDb (�106/ml

of hemolymph)

Total hemocytes FSDb (�106/ml

of hemolymph)

% Lamellocytes

F SDb

CgGAL4 8.3 F 5.43

(15)

25.0 F 2.89

(15)c15.5 F 5.18

(12)

e33CGAL4d 3.4 F 0.76

(10)

16.0 F 5.36

(13)cN/De

Lsp2GAL4 3.7 F 1.40

(15)

3.3 F 1.97

(15)fN/De

Note. The averages of total hemocyte counts excluding crystal cells were

presented. The number of larvae examined is indicated in parentheses.a Heterozygous larvae of mixed genotypes in culture.b Standard deviation.c Statistically significantdifferencesbetweenmutant andcontrol ( P b0.001).d Assayed at 288C.e Not determined.f Statistically insignificant differences between experimental and control.

L. Huang et al. / Developmental Biology 280 (2005) 407–420412

in the total hemocyte pool significantly increased in lwr and

Tl10B mutant larvae (Table 1 and Fig. 3). We also noticed

that most aggregated hemocytes in the hemolymph con-

sisted of lamellocytes, and that these aggregated masses

were partially to fully melanized in most cases (Fig. 2).

These observations strongly suggest that lamellocytes are

heavily involved in the formation of melanotic tumors in

lwr mutant larvae.

We also wonder whether the lwr mutant hemocytes

divide in circulation. We measured the mitotic indexes of

lwr mutant hemocytes as well as those of Tl10B and Canton-

S hemocytes as control. Cells in mitosis were identified

using anti-phospho-Histone H3 antibodies. The mitotic

index of lwr mutants was 10.4% in a total of 3109 cells

from five larvae, which was slightly higher than that of

Tl10B (7.1% in a total of 2515 cells from five larvae). On the

other hand, we did not observe any dividing hemocytes in

Canton-S (a total of 1344 cells from ten larvae). Based on

their sizes and morphologies, these dividing cells in lwr and

Tl10B mutants were plasmatocytes and prohemocytes, not

lamellocytes. Therefore, some hemocytes do indeed divide

in circulation in lwr mutants. However, the excess number

of mature lamellocytes in circulation is due to proliferation

of lamellocyte precursors, presumably in the lymph gland.

Taken together, we conclude that the effects of the lwr

mutation on hemocytes are similar to those of other known

hematopoietic mutants in the Tl pathway such as cact and

Tl10B (Qiu et al., 1998).

The overproduction of hemocytes is most likely due to the

loss of lwr function in the hematopoietic tissues

To determine whether the overproduction of hemocytes

can be attributed to the loss of lwr function in hematopoietic

Fig. 3. Plasmatocyte and lamellocyte populations in different genetic

backgrounds. Vertical bars represent the total hemocyte counts. Open space

in the vertical bar corresponds to plasmatocyte population and filled space

corresponds to lamellocyte population. The number of plasmatocytes and

lamellocytes was calculated based on the total hemocyte counts and

lamellocyte percentages. OR, Oregon-R (wild type); CS, Canton-S (wild

type); Tl10B, Tl10B/+; lwr, lwr4-3/lwr5; lwr dl, lwr4-3 dl1/lwr5 Df(2L)J4;

lwr Dif, lwr4-3 Dif2/lwr5 Df(2L)J4; lwr dl Dif, lwr4-3 Df(2L)J4/lwr5

Df(2L)TW119.

tissues, we expressed a dominant negative form of lwr

(lwrDN; see Materials and methods for the UAS-lwrDN

construct) in the lymph gland and hemocytes. Since the lwr

mutation exhibits pleiotropic effects (Apionishev et al.,

2001; Epps and Tanda, 1998), it is possible that the

increased number of hemocytes in lwr mutant larvae is

due to a secondary effect that might stimulate hemocyte

production. For this purpose, we used the GAL4/UAS

system (Brand and Perrimon, 1993). Two GAL4 drivers,

CgGAL4 and e33CGAL4, were chosen for this experiment

because they are known to induce expression of GAL4 in

the lymph glands and hemocytes (Asha et al., 2003;

Harrison et al., 1995). Although these drivers induce

GAL4 expression in multiple tissues, the common cell types

expressing GAL4 with these two drivers are hemocytes in

the lymph gland and in circulation. The e33CGAL4 driver

induces a UAS-GFP transgene in most cells in the lymph

gland at different expression levels, while the CgGAL4

promotes GFP expression in a subset of the cells in the

anterior lobes at a relatively consistent level. The overall

expression level of GFP by the e33CGAL4 is lower than

that of the CgGAL4 driver. Nevertheless, these GAL4

drivers exhibited similar effects on the total hemocyte

counts.

The CgGAL4 driver effectively increased the number of

hemocytes to a level that was higher than those of lwr and

Tl10B (Tables 1 and 2). The total hemocyte count was 25.0�106 per ml of hemolymph, which was statistically higher

than that of the corresponding control (8.3 � 106 per ml of

hemolymph, P b 0.001 in t test). The e33CGAL4 driver also

induced many hemocytes at 288C (16.0 � 106 per ml of

hemolymph), and this level was significantly different from

that of the corresponding heterozygous control (3.4� 106 ml

of hemolymph, P b 0.001 in t test). Since hemocytes in the

lymph gland and in circulation are the only common cell

types that express GAL4 with these drivers, it is most likely

that the loss of lwr function in hematopoietic tissues is

Page 7: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420 413

responsible for high hemocyte counts in lwr mutants. This

conclusion is also supported by the fact that a fat body

specific driver, Lsp2GAL4 (Cherbas et al., 2003), did not

show any significant effect on hemocyte counts when the

lwrDN allele was induced by this driver (Table 2).

In addition to the total hemocyte counts, the UAS-lwrDN/

CgGAL4 combination promoted lamellocyte production.

We observed that lamellocyte levels rose to 15.5% (Table 2).

This value was lower than that of lwr mutants, but higher

than that of Tl10B mutants. Furthermore, most hemocytes

expressing lwrDN showed nuclear localization of Dl protein

(Fig. 4B), which is characteristic of the lwr mutant

hemocytes (see below). These observations strongly suggest

that this lwrDN allele is very effective with the CgGAL4 in

mimicking effects of the lwr mutation on hemocyte

production in larvae. Thus, we conclude that the increase

in the total hemocyte counts in the lwr mutant background

is attributed to the loss of lwr function in the hematopoietic

tissues such as the lymph gland.

Fig. 4. Nuclear localization of Dorsal protein in Tl10B and lwr mutant hemocytes. (

type (labeled with WT) hemocytes were stained with anti-Dorsal monoclonal ant

DAPI to localize DNA (left column). Merged images (right column) clearly sho

(arrowheads), but not in wild type blood cells. Some cells (indicated with *) do no

Dorsal nuclear localization was examined in hemocytes expressing the lwrDN allele

UAS-GFP transgene (left panel). In the right panel, the cells with and without Dl

Two lamellocytes are seen overlapping in the center of the panels. Notice that Dl

cells. All photographs were taken at the same magnification, and the bar in white

Nuclear localization of the Dorsal protein was promoted in

lwr mutant hemocytes

How does the lwr mutation affect hemocyte production?

Because the hematopoietic defects of lwr mutants are

similar to those of dominant Tl mutations, Tl10B and Tl3

(Qiu et al., 1998), we investigated possible interactions

between lwr and the Tl signal transduction pathway.

Moreover, InBa, a human Cact homologue, is subject to

sumoylation (Desterro et al., 1998), which suggests that the

Cact protein may interact with the Lwr protein. Since both

dl and Dif are expressed in the hematopoietic tissues, we

used the entrance of Dl proteins as an indicator for Tl

signaling activity. At this moment, it is not yet clear whether

other Tl-like receptors are involved in hematopoiesis.

Hereby, the term Tl signaling in this report does not exclude

possible signaling inputs from other Tl-like receptors.

We observed nuclear localization of Dl protein in many

lwr mutant hemocytes as well as in Tl10B hemocytes (Fig.

A) Tl10B and lwr mutant (labeled with Tl10B and lwr, respectively) and wild

ibodies (middle column). The hemocytes were simultaneously stained with

w nuclear localization of Dl proteins in Tl10B and lwr mutant blood cells

t show nuclear localization of Dl protein in hemocytes of either mutant. (B)

with the CgGAL4 driver. The lwrDN expressing cells were visualized with a

nuclear localization are labeled with arrowheads and asterisks, respectively.

nuclear localization is seen in GFP-positive cells, but not in GFP-negative

in the top left panel corresponds to 20 Am.

Page 8: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420414

4A). In lwr mutants, nearly half of hemocytes (46.9% in a

total of 1960 cells from five larvae) showed accumulation of

Dl proteins in the nuclei, which was comparable to that of

Tl10B mutants (48.2% in a total of 2699 cells from five

larvae). In contrast, nuclear localization of Dl protein was

observed in only a few control heterozygous and wild type

hemocytes, e.g., 3.6% in a total of 1291 cells from 10Canton-

S larvae. These observations indicate that the activation of the

Tl pathway is a consequence of the loss of lwr function,

which is manifested as abnormal production of larval

hemocytes. Although some small lamellocytes were positive

for Dl nuclear staining, little nuclear localization of Dl protein

was observed in large matured lamellocytes of lwr and Tl10B

mutants. Thus, there is not a mechanism to allow entrance of

Dl proteins to the nucleus in mature lamellocytes.

The dorsal and Dif mutations are suppressors of the lwr

mutations in hematopoiesis

To further investigate the relationship between the lwr

mutation and Tl signaling, we examined genetic interactions

of lwr with dl and Dif single mutants as well as with dl Dif

double mutants. The rationale here is that genetic inter-

actions between lwr and dl as well as Dif would be clearly

detected if lwr function modulates signaling from Tl

receptor and possibly from other Tl-like receptors. We

describe the results of these three combinations separately in

the following sections because the dl and the Dif mutations

showed different effects on the hematopoietic defects of the

lwr mutation. Nonetheless, the results described below

indicate that the majority of lwr’s mutant effects on larval

hematopoiesis are manifested through the Tl signal trans-

duction pathway, which agrees with the results of our

immunohistochemical study (see above).

The dorsal mutation suppresses the production of

plasmatocytes in the lwr mutant background

We created a double mutant combination of lwr with

dl [lwr4-3 dl1/lwr5 Df(2L)J4]. The total hemocyte counts

and lamellocyte percentages were measured (Table 1). The

total hemocyte counts were significantly reduced in the

lwr dl double mutant background (P b 0.001 in

Bonferroni’s test). The numbers of plasmatocytes and

lamellocytes were then calculated based on the total

hemocyte counts and lamellocyte percentages. We noticed

that plasmatocyte counts were reduced by approximately

50% in the double mutant combination. Interestingly, the

production of lamellocytes was not overly affected in the

same mutant background (Fig. 3). which indicates that dl

plays a minor role in lamellocyte production.

Although the production of plasmatocytes was sup-

pressed by the complete loss of dl function, this suppression

was not absolute, suggesting that Dif may contribute equally

to larval plasmatocyte production. If Dif only played a

minor role in this process, the lwr mutation would affect

plasmatocyte production via another pathway besides the Tl

pathway. Note that the latter hypothesis was proven to be

very unlikely (see below).

The Dif mutation suppressed the production of both

plasmatocytes and lamellocytes in the lwr mutant

background

To examine the effect of Dif in the lwr mutant

background, we created a double mutant combination of

lwr and Dif [lwr4-3 Dif2/lwr5 Df(2L)J4] and measured the

total hemocyte counts and lamellocyte percentages (Table

1). The loss of Dif function in the lwr background

significantly reduced total hemocyte counts (P b 0.001 in

Bonferroni’s test), and the effects were observed on both

plasmatocyte and lamellocyte levels. Plasmatocyte popula-

tion was reduced to half the level caused by the lwr

mutation, while the number of lamellocytes was similar to

that found in wild type larvae (Fig. 3).

Similar to what we observed in the lwr dl double mutants,

the reduction of the plasmatocyte population due to the loss

of Dif function in the lwr background was approximately

50%. This result strongly suggests, in conjunction with the

results of the lwr dl double mutants, that the increase of

hemocytes observed in the lwr mutation is mediated by the

functions of both dl and Dif (also see below).

In contrast to the lwr dl double mutant, the lamellocyte

population was considerably affected by the introduction of

the Dif mutation into the lwr mutant background, and the

number of lamellocytes observed in the lwr Dif double

mutants was almost identical to those in wild type larvae

(Fig. 3). Therefore, lamellocyte production is primarily

controlled by Dif function when the Tl pathway is activated.

This in turn suggests that dl and Dif play different roles in

hematopoiesis when it is stimulated by the lwr mutation.

Loss of both dl and Dif functions totally diminished the

effects of the lwr mutation on hemocyte production

To obtain a more conclusive answer as to whether the

effects of the lwr mutation are observed mostly through Rel-

related proteins, Dl and Dif, we examined lwr dl Dif triple

mutants [lwr4-3 Df(2L)J4/lwr5 Df(2L)TW119]. We observed

that the loss of both gene functions almost completely

cancelled the effects of the lwr mutation on hematopoiesis.

We eliminated the functions of dl and Dif by combining

the deficiencies Df(2L)J4 and Df(2L)TW119 (see Materials

and methods). Total hemocyte counts of lwr5 Df(2L)J4/

lwr4-3 Df(2L)TW119 heterozygotes were significantly

reduced from those of the lwr single mutants (P b 0.001

in Bonferroni’s test) and were indistinguishable from those

in wild type larvae as well as Df(2L)J4/ Df(2L)TW119

heterozygotes (Table 1). The number of lamellocytes was

almost identical to those of wild type larvae (Fig. 3). These

results indicate that the hematopoietic defects of the lwr

mutation are manifested through dl and Dif function.

Page 9: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420 415

The loss of both dl and Dif functions with and without

the lwr mutation did not lead to complete loss of hemocytes.

This observation agrees with the fact that loss-of-function

mutations of the Tl gene did not eliminate all hemocytes

completely (Qiu et al., 1998). Low levels of hemocytes in

these mutant backgrounds can be explained by hemocyte

production using other pathways such as JAK/STAT and

Ras, which are known to be involved in hematopoiesis

(Asha et al., 2003; Harrison et al., 1995; Luo et al., 1997).

Therefore, these observations do not rule out the importance

of dl and Dif functions in hematopoiesis.

dl and Dif possess different functions in hematopoiesis

In order to obtain additional evidence that supports

different but overlapping roles of dl and Dif in hematopoi-

esis, we overexpressed UAS-dl and UAS-Dif transgenes in

the lymph glands and hemocytes using the CgGAL4 driver

(Table 3). We observed that Dif promoted lamellocyte

production more effectively than dl did, which shows good

agreement with our genetic analysis (see above).

Since the CgGAL4 driver expresses the GAL4 tran-

scription factor in the fat body, we also used the Lsp2GAL4

driver, which is fat body specific (Cherbas et al., 2003), to

express the UAS constructs used in this study. The

combinations of all UAS transgenes with the Lsp2GAL4

serve as a control to assess any possible additional effect of

the CgGAL4 driver. In all cases examined, while the

differences between controls and experimental sets were

statistically significant in some cases (Table 3), total

hemocyte counts fell in the range of wild type, Oregon-R

and Canton-S (Table 1). Therefore, the results with the

CgGAL4 driver, which are described in the following

paragraphs, are most likely to represent the effects of these

genes in the hematopoietic tissues, the lymph gland and

hemocytes.

Table 3

Effects of dl, Dif, and Tl10B transgenes on larval hemocytes

GAL4 driver UAS transgene Controla

Total hemocytes FSDb (�106/ml of

hemolymph)

CgGAL4 dorsal 2.6 F 1.18 (35)

Dif 2.3 F 1.25 (35)

dorsal + Dif 1.9 F 0.78 (35)

Toll10B 1.8 F 0.46 (35)

Lsp2GAL4 dorsal 1.7 F 0.69 (12)

Dif 3.6 F 1.13 (11)

dorsal + Dif 1.7 F 0.44 (35)

Toll10B 1.9 F 0.61 (35)

Note. The averages of total hemocyte counts excluding crystal cells were presenta Heterozygous larvae of mixed genotypes in culture.b Standard deviation.c Statistically significant differences between mutant and control ( P b 0.001).d Statistically insignificant differences between experimental and control.e Not determined.

Both dl and Dif exhibited significant increases in total

hemocyte counts when they were overexpressed by the

CgGAL4 driver. The average total hemocyte counts of these

UAS/GAL4 combinations were statistically different from

those of the corresponding heterozygous controls (P b 0.001

in t test). Interestingly, overexpression of Dif produced more

hemocytes than dl. After dividing the total hemocyte

population into plasmatocytes and lamellocytes, we noticed

that plasmatocyte counts increased to levels similar to those

of lwr and Tl10B mutants in both UAS-dl/CgGAL4 and UAS-

Dif/CgGAL4 combinations (Figs. 3 and 5). Unlike the

plasmatocyte population, lamellocytes responded differently

(Fig. 5). Overexpression of dl showed no effect on

lamellocyte production. In contrast, overexpression of Dif

promoted lamellocyte production and its effect was similar to

that of a Tl10B transgene. These results indicate that dl andDif

share a similar function in plasmatocyte production, and that

Dif is likely to be a sole factor for lamellocyte production in

the Tl pathway.

Dif function can replace dl in hematopoiesis

To investigate how coordinately dl and Dif function in

hematopoiesis, we examined effects on hemocyte produc-

tion by overexpressing Tl10B and both dl and Dif simulta-

neously. We found that dl may be dispensable in the

production of both plasmatocytes and lamellocytes when

there were enough Dif proteins around. Furthermore, the

activation of Tl signaling by Tl10B, a constitutively active

form of the Tl receptor, showed an additional effect on

plasmatocyte production compared to those by the simulta-

neous expression of dl and Dif.

We overexpressed dl and Dif simultaneously with the

CgGAL4 driver and estimated the total hemocyte number

and the proportion of lamellocytes in the total hemocyte

population (Table 3). Even though dl and Dif showed

Experimental

Total hemocytes FSDb (�106/ml of

hemolymph)

% Lamellocytes F SDb

18.0 F 4.53 (35)c 1.7 F 0.55 (15)

30.8 F 13.5 (35)c 11.7 F 1.58 (15)

26.0 F 5.49 (35)c 19.6 F 2.87 (15)

39.6 F 9.62 (35)c 11.4 F 4.26 (15)

1.4 F 0.59 (12)d N/De

1.6 F 0.96 (12)c N/De

3.3 F 0.69 (35)c N/De

3.1 F 0.94 (35)c N/De

ed. The number of larvae examined is indicated in parentheses.

Page 10: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

Fig. 5. Plasmatocyte and lamellocyte populations induced by different UAS

transgenes with the CgGAL4. Vertical bars represent the total hemocyte

counts. Open space in the vertical bar corresponds to plasmatocyte

population and filled space corresponds to lamellocyte population. The

number of plasmatocytes and lamellocytes was calculated based on the total

hemocyte counts and lamellocyte percentages.

L. Huang et al. / Developmental Biology 280 (2005) 407–420416

significant effects on hemocyte production when they

were individually overexpressed, they did not show any

synergistic effect when they were induced at the same

time. We applied ANOVA to analyze our date set

presented in Table 3, and Bonferroni’s multiple compar-

ison test was used to measure the significance of the

differences among the UAS transgene combinations.

When both genes were expressed together, the total

hemocyte counts did not differ from those when Dif was

overexpressed by the CgGAL4 driver, but were statisti-

cally higher than those when dl was overexpressed by

the same driver (P b 0.001 in Bonferroni’s test). Thus,

Dif is sufficient to represent the effect of the dl and Dif

double expression combination, and dl did not suppress

the effect of Dif.

As far as lamellocyte production is concerned, the

combination of dl and Dif showed the highest lamellocyte

estimate among all constructs tested including UAS-Tl10B.

Although a statistical test could not be applied, the differ-

ences among the constructs seemed to be marginal,

indicating that the effect of dl on lamellocyte production

may be small, if there is any. Taken together, dl only plays a

minimal role in hematopoiesis when both dl and Dif are

highly induced.

In order to verify whether the dl-Dif double combina-

tion represents the activation of the Tl pathway, we

overexpressed a UAS-Tl10B transgene with the CgGAL4

driver. While lamellocyte production appeared to be very

similar to that of a dl–Dif double combination, the

overexpression of Tl10B exhibited the most pronounced

effect on plasmatocyte production among all the combi-

nations used in this study (Fig. 5). The results indicate that

there are abundant Dl and Dif proteins in the CgGAL4

expressing cells and that these transcription factors can

fully respond to the activation of the Tl pathway, i.e., the

overexpression of Tl10B. It is also possible that the

activation of the Tl receptor may stimulate plasmatocyte

production, in part bypassing the Dl and Dif transcription

factors. This possible bypass indicates that the Tl receptor

might use a different set of transducers and transcription

factors to control plasmatocyte production. Alternatively,

the differences might be due to the different levels of UAS

transgene expression.

Discussion

We have demonstrated that the mutation of the lwr gene,

which encodes a SUMO-conjugating enzyme, causes the

activation of Rel-related proteins, Dl and Dif, and leads to

overproduction of larval hemocytes in D. melanogaster. Our

genetic analysis also indicates that dl and Dif have different

functions in larval hematopoiesis.

The lwr mutation leads to the activation of Rel-related

proteins in Drosophila larval hemocytes

Our immunochemical and genetic analyses indicate that

the lwr mutation results in the activation of Dl and Dif

primarily in plasmatocyte and prohemocytes, leading to

overproliferation of plasmatocytes and lamellocytes in

larvae. Dl nuclear localization in the lwr mutant hemocytes

was observed as frequently as those in the Tl10B mutant

hemocytes (Fig. 4A). The high hemocyte counts of the lwr

mutant larvae were almost completely suppressed by

removing both dl and Dif functions from the lwr mutant

background (Table 1). Therefore, the present study proposes

that the wild type lwr function negatively regulates the

activity of the Tl pathway and/or the pathways of Tl-like

genes.

What is a possible molecular mechanism of this

regulation? The Lwr protein can regulate activities of its

target proteins by conjugating SUMO molecules. For

example, the human Cact homologue, IjBa, is known to

be sumoylated, and the sumoylated IjBa molecules were

shown to be resistant to ubiquitination because the

ubiquitination sites on IjBa are occupied by SUMO

molecules (Desterro et al., 1998). In other words, sumoy-

lation stabilizes IjBa, which in turn represses NF-nBactivity. Alternatively, their physical associations alone can

control physiological functions of Lwr’s targets. Recently,

the Paired-like homeobox protein Vsx-1 was reported to

physically interact with Ubc9 (the vertebrate Lwr homo-

logue) and to require Ubc9 function for Vsx-1’s nuclear

localization. However, sumoylation of the Vsx-1 protein

was not detected (Kurtzman and Schechter, 2001). In this

case, physical association with Ubc9 itself is a key for Vsx-1

function.

In the Tl (or Tl-like) pathway of Drosophila, the above

two different mechanisms are possible (Fig. 6A, B). The

Dl protein was shown to physically interact with Lwr and

to be sumoylated in a transfection experiment using

Page 11: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

Fig. 6. Models of the Lwr–Cact interaction. These models show how the

Lwr protein possibly interacts with its targets using the Cact protein as

an example. Model in (A) represents interaction via SUMO conjugation,

and Model in (B) represents simple physical interference. For simplicity,

either Dl or Dif is depicted in each model, and each model can be

applied to both Dl and Dif. In lwr mutants, Cact becomes more

susceptible for ubiquitin-mediated degradation in the cytoplasm (two

paths in the middle), which leads to the activation of Dl and Dif

downstream genes in the nucleus. Letter K on the Cact protein represents

a lysine residue that can be modified by both SUMO and ubiquitin.

Panel C shows different roles of Dl and Dif in Drosophila larval

hematopoiesis.

L. Huang et al. / Developmental Biology 280 (2005) 407–420 417

Schneider L2 cells. Sumoylated Dl proteins dissociated

from Cact and entered the nucleus, where they exhibited

an increased level of transactivation (Bhaskar et al., 2000,

2002). However, this model does not fit our observations

that showed a higher incidence of Dl nuclear localization

in lwr mutant hemocytes than in wild type hemocytes

(Fig. 4A).

Another possible target protein is Cact (Fig. 6). As in

IjBa, the Cact protein was shown to physically associate

with Lwr (Bhaskar et al., 2000). Unlike IjBa, sumoylated

Cact proteins were not detected in the same study. However,

physical association of Lwr and Cact might stabilize Cact

proteins in the cytoplasm by counteracting ubiquitination or

phosphorylation, a prerequisite for ubiquitination of Cact

(Reach et al., 1996). In this scenario, the loss of lwr function

would make Cact be susceptible to degradation, which

results in migration of Dl and Dif to the nucleus (Fig. 6B).

Other components in the Tl pathway could be regulated

similarly, although they have not been reported to interact

with Lwr. These possibilities are under study in our

laboratory.

Loss of lwr function does not affect nuclear transport of Dl

protein in hemocytes

Our previous study demonstrated that lwr function was

required for the Ran-dependent nuclear transport system

(Epps and Tanda, 1998). This raises a question of how

efficiently Dl and Dif proteins are transported to the nucleus

in lwr mutant hemocytes. Our observations indicate that

nuclear import of Dl and Dif was not overly impaired. These

contrary observations can be explained by functional

redundancy of lwr with 25 other Drosophila E2 enzymes.

Since functions of most E2 enzymes have not been clearly

defined, some of them may be SUMO conjugases. This

possibility was also suggested in the embryo, particularly in

the posterior half of the embryo in the previous study (Epps

and Tanda, 1998). Alternatively, the residual lwr function

may be sufficient to mediate nuclear import of Dl proteins

because we used hypomorphic lwr alleles in this study. In

any case, a possible negative effect on nuclear transport

appeared minimal in lwr mutant hemocytes.

Dif and dl play different roles in hemocyte production

The loss of dl function in the lwr mutant background

resulted in a reduction of plasmatocyte, not lamellocyte

counts (Table 1), and the overexprssion of dl with the

CgGAL4 driver stimulated plasmatocyte production only

(Table 3). Therefore, the role of dl in hematopoiesis is

limited to plasmatocyte production (Fig. 6C). Although dl

plays a role in plasmatocyte production in the wild type

background, the contribution of dl to plasmatocyte produc-

tion seems minimal when Dif is overexpressed (Table 3). In

other words, Dif can replace dl function in Drosophila

hematopoiesis.

On the other hand, Dif is capable of stimulating the

production of both plasmatocytes and lamellocytes (Fig.

6C). Because the loss of Dif function in the lwr mutant

background led the lamellocyte population to a level very

close to those of wild type strains (Table 1), Dif is most

likely to play an essential and sole role in lamellocyte

production when the Tl signaling is activated by the lwr

mutation. Furthermore, when Dif was overexpressed by the

CgGAL4, the levels of plasmatocytes and lamellocytes were

similar to those reached when Tl10B was overexpressed by

the same driver. Taken together, we conclude that Dif is

necessary and sufficient to promote the production of

plasmatocytes and lamellocytes in larvae in the Tl (Tl-like)

pathway. This also means that Dl-responsive genes can be

regulated by Dif.

We hypothesize that the genes for plasmatocyte produc-

tion can be divided into at least two classes. The first class

consists of genes regulated primarily by Dl. The genes in

this class can be stimulated by Dif in the absence of Dl. It is

known that Dl proteins bind to more strictly defined nB sites

than Dif, and that Dif can bind more broadly to nBconsensus sequences including Dl-binding sites (Engstrom

Page 12: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420418

et al., 1993; Gross et al., 1996; Han and Ip, 1999; Petersen

et al., 1995). Several humoral immunity genes were found

to belong to this class (Engstrom et al., 1993; Gross et al.,

1996). Thus, it is possible that such genes exist among

genes essential for plasmatocyte production. The other class

includes genes regulated only by Dif. In these genes,

consensus nB sites are probably responsible for their

expression via the Dif protein.

The Dl protein can function as a repressor in a context-

dependent manner (Huang et al., 1993; Jiang et al., 1993;

Kirov et al., 1993; Markstein et al., 2002). A good example

in the Drosophila immunity is the Cecropin gene, whose

expression depends heavily on Dif (Engstrom et al., 1993).

Co-expression of Dl expression along with Dif strongly

repressed the expression of the Cecropin gene. However, we

did not observe any significant dominant negative effect of

dl on plasmatocyte production in our overexpression

experiments (Table 3). At the same time, we did not

observe any synergistic effect on plasmatocyte production

when both dl and Dif were co-expressed (Table 3).

Therefore, regulatory mechanisms of plasmatocyte-specific

genes are not so complex.

Plasmatocytes can proliferate upon the activation of Dl and

Dif

The collagen IV (Cg25C) gene, whose promoter was

used for the CgGAL4 driver, is expressed in embryonic and

larval hemocytes (Yasothornsrikul et al., 1997). These larval

hemocytes are most likely plasmatocytes, although the

authors of the study did not distinguish plasmatocytes and

prohemocytes. Nonetheless, our results show that these

Cg25C-expressing hemocytes are capable of proliferating

and differentiating into plasmatocytes when Dl and Dif is

activated.

It was surprising that the CgGAL4 driver induced a

relatively large number of lamellocytes because this driver

is not capable of inducing GAL4 in lamellocytes (Asha et

al., 2003) and because the collagen IV gene is expressed

primarily in plasmatocytes. This can be interpreted in two

different ways. The first one is that the Cg25C gene is

expressed in lamellocyte precursors in the lymph gland,

where these precursor cells can divide. In addition to

different cell types distinguished by their morphologies, the

cells in the lymph gland can be classified based on

expression of different genes. For example, the Posterior

Signaling Center, which is important for crystal cell differ-

entiation, was defined as Serrate-expressing cells in the first

lobe of the lymph gland (Lebestky et al., 2003). Thus, it is

conceivable that the Cg25C gene might be expressed in the

cells that have not been clearly defined in the lymph gland.

The second interpretation is that plasmatocytes can differ-

entiate into lamellocytes when Dif are highly activated. This

interpretation supports a classical view of lamellocyte

differentiation (Rizki and Rizki, 1984). Further investiga-

tions are necessary to answer these questions.

Acknowledgments

The authors thank C.R. Dearolf, J.A. Hoffmann, Y.T. Ip,

L. Wu, and The Bloomington Drosophila Stock Center for

valuable stocks, Developmental Studies Hybridoma Bank

for antibodies, E. Baereke for the use of his histology set-up,

Y.T. Ip, S. Govind, and M. Meister for constructive

discussion. Special appreciation goes to M.R. Van Doren

for technical assistance and valuable comments on this

manuscript, to M.E. Van Doren for editorial work on the

manuscript, and to Y. Shen and T. Bullock for excellent

technical assistance. This work was supported in part by a

grant from the National Institutes of Health to S.T. and by

Ohio University.

References

Anderson, K.V., Jurgens, G., Nusslein-Volhard, C., 1985. Establishment of

dorsal–ventral polarity in the Drosophila embryo: genetic studies on the

role of the Toll gene product. Cell 42, 779–789.

Apionishev, S., Malhotra, D., Raghavachari, S., Tanda, S., Rasooly, R.S.,

2001. The Drosophila UBC9 homologue lesswright mediates the

disjunction of homologues in meiosis I. Genes Cells 6, 215–224.

Asha, H., Nagy, I., Kovacs, G., Stetson, D., Ando, I., Dearolf, C.R., 2003.

Analysis of Ras-induced overproliferation in Drosophila hemocytes.

Genetics 163, 203–215.

Baldwin Jr., A.S., 1996. The NF-kappa B and I kappa B proteins: new

discoveries and insights. Annu. Rev. Immunol. 14, 649–683.

Bhaskar, V., Valentine, S.A., Courey, A.J., 2000. A functional interaction

between dorsal and components of the Smt3 conjugation machinery.

J. Biol. Chem. 275, 4033–4040.

Bhaskar, V., Smith, M., Courey, A.J., 2002. Conjugation of Smt3 to dorsal

may potentiate the Drosophila immune response. Mol. Cell. Biol. 22,

492–504.

Bos, J.L., 1989. ras oncogenes in human cancer: a review. Cancer Res. 49,

4682–4689.

Brand, A.H., Perrimon, N., 1993. Targeted gene expression as a means of

altering cell fates and generating dominant phenotypes. Development

118, 401–415.

Braun, A., Lemaitre, B., Lanot, R., Zachary, D., Meister, M., 1997.

Drosophila immunity: analysis of larval hemocytes by P-element-

mediated enhancer trap. Genetics 147, 623–634.

Cherbas, L., Hu, X., Zhimulev, I., Belyaeva, E., Cherbas, P., 2003. EcR

isoforms in Drosophila: testing tissue-specific requirements by targeted

blockade and rescue. Development 130, 271–284.

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.

Desterro, J.M., Rodriguez, M.S., Hay, R.T., 1998. SUMO-1 modifica-

tion of IkappaBalpha inhibits NF-kappaB activation. Mol. Cell 2,

233–239.

Dushay, M.S., Asling, B., Hultmark, D., 1996. Origins of immunity: Relish,

a compound Rel-like gene in the antibacterial defense of Drosophila.

Proc. Natl. Acad. Sci. U. S. A. 93, 10343–10347.

Duvic, B., Hoffmann, J.A., Meister, M., Royet, J., 2002. Notch signaling

controls lineage specification during Drosophila larval hematopoiesis.

Curr. Biol. 12, 1923–1927.

Engstrom, Y., Kadalayil, L., Sun, S.C., Samakovlis, C., Hultmark, D., Faye,

I., 1993. kappa B-like motifs regulate the induction of immune genes in

Drosophila. J. Mol. Biol. 232, 327–333.

Epps, J.L., Tanda, S., 1998. The Drosophila semushi mutation blocks

nuclear import of bicoid during embryogenesis. Curr. Biol. 8,

1277–1280.

Page 13: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420 419

Evans, C.J., Hartenstein, V., Banerjee, U., 2003. Thicker than blood:

conserved mechanisms in Drosophila and vertebrate hematopoiesis.

Dev. Cell 5, 673–690.

Geisler, R., Bergmann, A., Hiromi, Y., Nusslein-Volhard, C., 1992. cactus,

a gene involved in dorsoventral pattern formation of Drosophila, is

related to the I kappa B gene family of vertebrates. Cell 71, 613–621.

Ghosh, S., May, M.J., Kopp, E.B., 1998. NF-kappa B and Rel proteins:

evolutionarily conserved mediators of immune responses. Annu. Rev.

Immunol. 16, 225–260.

Gross, I., Georgel, P., Kappler, C., Reichhart, J.M., Hoffmann, J.A., 1996.

Drosophila immunity: a comparative analysis of the Rel proteins dorsal

and Dif in the induction of the genes encoding diptericin and cecropin.

Nucleic Acids Res. 24, 1238–1245.

Han, Z.S., Ip, Y.T., 1999. Interaction and specificity of Rel-related proteins

in regulating Drosophila immunity gene expression. J. Biol. Chem.

274, 21355–21361.

Harrison, D.A., Binari, R., Nahreini, T.S., Gilman, M., Perrimon, N., 1995.

Activation of a Drosophila Janus kinase (JAK) causes hematopoietic

neoplasia and developmental defects. EMBO J. 14, 2857–2865.

Hatada, E.N., Krappmann, D., Scheidereit, C., 2000. NF-kappaB and the

innate immune response. Curr. Opin. Immunol. 12, 52–58.

Hoffmann, J.A., Reichhart, J.M., 2002. Drosophila innate immunity: an

evolutionary perspective. Nat. Immunol. 3, 121–126.

Hoffmann, J.A., Kafatos, F.C., Janeway, C.A., Ezekowitz, R.A., 1999.

Phylogenetic perspectives in innate immunity. Science 284, 1313–1318.

Hu, X., Yagi, Y., Tanji, T., Zhou, S., Ip, Y.T., 2004. Multimerization and

interaction of Toll and Spatzle in Drosophila. Proc. Natl. Acad. Sci.

U. S. A. 101, 9369–9374.

Huang, J.D., Schwyter, D.H., Shirokawa, J.M., Courey, A.J., 1993. The

interplay between multiple enhancer and silencer elements defines the

pattern of decapentaplegic expression. Genes Dev. 7, 694–704.

Hultmark, D., 1993. Immune reactions in Drosophila and other insects: a

model for innate immunity. Trends Genet. 9, 178–183.

Ip, Y.T., Reach, M., Engstrom, Y., Kadalayil, L., Cai, H., Gonzalez-Crespo,

S., Tatei, K., Levine, M., 1993. Dif, a dorsal-related gene that mediates

an immune response in Drosophila. Cell 75, 753–763.

Jiang, J., Cai, H., Zhou, Q., Levine, M., 1993. Conversion of a dorsal-

dependent silencer into an enhancer: evidence for dorsal corepressors.

EMBO J. 12, 3201–3209.

Kidd, S., 1992. Characterization of the Drosophila cactus locus and

analysis of interactions between cactus and dorsal proteins. Cell 71,

623–635.

Kimbrell, D.A., Beutler, B., 2001. The evolution and genetics of innate

immunity. Nat. Rev., Genet. 2, 256–267.

Kirov, N., Zhelnin, L., Shah, J., Rushlow, C., 1993. Conversion of a

silencer into an enhancer: evidence for a co-repressor in dorsal-

mediated repression in Drosophila. EMBO J. 12, 3193–3199.

Kisseleva, T., Bhattacharya, S., Braunstein, J., Schindler, C.W., 2002.

Signaling through the JAK/STAT pathway, recent advances and future

challenges. Gene 285, 1–24.

Kurtzman, A.L., Schechter, N., 2001. Ubc9 interacts with a nuclear

localization signal and mediates nuclear localization of the paired-like

homeobox protein Vsx-1 independent of SUMO-1 modification. Proc.

Natl. Acad. Sci. U. S. A. 98, 5602–5607.

Lanot, R., Zachary, D., Holder, F., Meister, M., 2001. Postembryonic

hematopoiesis in Drosophila. Dev. Biol. 230, 243–257.

Lebestky, T., Chang, T., Hartenstein, V., Banerjee, U., 2000. Specification

of Drosophila hematopoietic lineage by conserved transcription factors.

Science 288, 146–149.

Lebestky, T., Jung, S.H., Banerjee, U., 2003. A Serrate-expressing signaling

center controls Drosophila hematopoiesis. Genes Dev 17, 348–353.

Luo, H., Rose, P., Barber, D., Hanratty, W.P., Lee, S., Roberts, T.M.,

D’Andrea, A.D., Dearolf, C.R., 1997. Mutation in the Jak kinase JH2

domain hyperactivates Drosophila and mammalian Jak-Stat pathways.

Mol. Cell. Biol. 17, 1562–1571.

Markstein, M., Markstein, P., Markstein, V., Levine, M.S., 2002. Genome-

wide analysis of clustered Dorsal binding sites identifies putative target

genes in the Drosophila embryo. Proc. Natl. Acad. Sci. U. S. A. 99,

763–768.

McCormick, F., 1994. Activators and effectors of ras p21 proteins. Curr.

Opin. Genet. Dev. 4, 71–76.

Meister, M., Lemaitre, B., Hoffmann, J.A., 1997. Antimicrobial peptide

defense in Drosophila. Bioessays 19, 1019–1026.

Melchior, F., 2000. SUMO—nonclassical ubiquitin. Annu. Rev. Cell Dev.

Biol. 16, 591–626.

Meng, X., Khanuja, B.S., Ip, Y.T., 1999. Toll receptor-mediated Drosophila

immune response requires Dif, an NF-kappaB factor. Genes Dev. 13,

792–797.

Muller, S., Hoege, C., Pyrowolakis, G., Jentsch, S., 2001. SUMO,

ubiquitin’s mysterious cousin. Nat. Rev., Mol. Cell Biol. 2, 202–210.

Nusslein-Volhard, C., Lohs-Schardin, M., Sander, K., Cremer, C., 1980. A

dorso-ventral shift of embryonic primordia in a new maternal-effect

mutant of Drosophila. Nature 283, 474–476.

O’Shea, J.J., Gadina, M., Schreiber, R.D., 2002. Cytokine signaling in

2002: new surprises in the Jak/Stat pathway. Cell 109, S121–S131

(Suppl.).

Petersen, U.M., Bjorklund, G., Ip, Y.T., Engstrom, Y., 1995. The

dorsal-related immunity factor, Dif, is a sequence-specific trans-

activator of Drosophila Cecropin gene expression. EMBO J. 14,

3146–3158.

Presnell, J., Schreibman, M., 1997. Animal Tissue Techniques. The Johns

Hopkins Univ. Press, Baltimore.

Qiu, P., Pan, P.C., Govind, S., 1998. A role for the Drosophila Toll/Cactus

pathway in larval hematopoiesis. Development 125, 1909–1920.

Reach, M., Galindo, R.L., Towb, P., Allen, J.L., Karin, M., Wasserman,

S.A., 1996. A gradient of cactus protein degradation establishes

dorsoventral polarity in the Drosophila embryo. Dev. Biol. 180,

353–364.

Rizki, T., 1957. Alterations in the haemocyte population of Drosophila

melanogaster. J. Morphol. 100, 437–458.

Rizki, T.M., 1978. The circulatory system and associated cells and tissues.

In: Ashburner, M., Wright, T.R.F. (Eds.), The Genetics and Biology of

Drosophila, vol. 2b. Academic Press, New York, pp. 397–452.

Rizki, R.M., Rizki, T.M., 1974. Basement membrane abnormalities

in melanotic tumor formation of Drosophila. Experientia 30,

543–546.

Rizki, T., Rizki, R., 1984. The cellular defense system of Drosophila

melanogaster. In: King, R., Akai, H. (Eds.), Insect Ultrastructure, vol.

2. Plenum Press, New York, pp. 579–604.

Robertson, H.M., Preston, C.R., Phillis, R.W., Johnson-Schlitz, D.M.,

Benz, W.K., Engels, W.R., 1988. A stable genomic source of P

element transposase in Drosophila melanogaster. Genetics 118,

461–470.

Rushlow, C.A., Han, K., Manley, J.L., Levine, M., 1989. The graded

distribution of the dorsal morphogen is initiated by selective nuclear

transport in Drosophila. Cell 59, 1165–1177.

Schmidt, D., Muller, S., 2003. PIAS/SUMO: new partners in transcriptional

regulation. Cell. Mol. Life Sci. 60, 2561–2574.

Schupbach, T., 1987. Germ line and soma cooperate during oogenesis to

establish the dorsoventral pattern of egg shell and embryo in Drosophila

melanogaster. Cell 49, 699–707.

Sen, R., Baltimore, D., 1986. Multiple nuclear factors interact with the

immunoglobulin enhancer sequences. Cell 46, 705–716.

Shuai, K., Liu, B., 2003. Regulation of JAK-STAT signalling in the immune

system. Nat. Rev., Immunol. 3, 900–911.

Silverman, N., Maniatis, T., 2001. NF-kappaB signaling pathways in

mammalian and insect innate immunity. Genes Dev. 15, 2321–2342.

Sinenko, S.A., Kim, E.K., Wynn, R., Manfruelli, P., Ando, I., Wharton,

K.A., Perrimon, N., Mathey-Prevot, B., 2004. Yantar, a conserved

arginine-rich protein is involved in Drosophila hemocyte development.

Dev. Biol. 273, 48–62.

Sorrentino, R.P., Carton, Y., Govind, S., 2002. Cellular immune response to

parasite infection in the Drosophila lymph gland is developmentally

regulated. Dev. Biol. 243, 65–80.

Page 14: The lesswright mutation activates Rel-related proteins, leading to overproduction of larval hemocytes in Drosophila melanogaster

L. Huang et al. / Developmental Biology 280 (2005) 407–420420

Spradling,A, 1986. P element-mediated transformation. In: Roberts,D. (Ed.),

Drosophila, A Practical Approach. IRL Press, Oxford, pp. 175–197.

Steward, R., 1987. Dorsal, an embryonic polarity gene in Drosophila, is

homologous to the vertebrate proto-oncogene, c-rel. Science 238,

692–694.

Sullivan, W., Ashburner, M., Hawley, R., 2000. Drosophila Protocols. Cold

Spring Harbor Laboratory Press, Cold Spring Harbor.

Sun, X., Huang, L., Van Doren, M.R., Tanda, S., 2003. Isolation of

amorphic alleles of the lesswright gene by P element-mediated male

recombination in Drosophila melanogaster. DIS 86, 79–83.

Tashiro, K., Pando, M.P., Kanegae, Y., Wamsley, P.M., Inoue, S., Verma,

I.M., 1997. Direct involvement of the ubiquitin-conjugating enzyme

Ubc9/Hus5 in the degradation of IkappaBalpha. Proc. Natl. Acad. Sci.

U. S. A. 94, 7862–7867.

Tzou, P., De Gregorio, E., Lemaitre, B., 2002. How Drosophila combats

microbial infection: a model to study innate immunity and host–

pathogen interactions. Curr. Opin. Microbiol. 5, 102–110.

Ward, A.C., Touw, I., Yoshimura, A., 2000. The Jak-Stat pathway in normal

and perturbed hematopoiesis. Blood 95, 19–29.

Yasothornsrikul, S., Davis, W.J., Cramer, G., Kimbrell, D.A., Dearolf, C.R.,

1997. Viking: identification and characterization of a second type IV

collagen in Drosophila. Gene 198, 17–25.

Yeh, E.T., Gong, L., Kamitani, T., 2000. Ubiquitin-like proteins: new wines

in new bottles. Gene 248, 1–14.