Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

a r t ic l e s

Toll-6 and Toll-7 function as neurotrophin receptors in


the Drosophila melanogaster CNS
Graham McIlroy1,3, Istvan Foldi1,3, Jukka Aurikko2, Jill S Wentzell1, Mei Ann Lim1, Janine C Fenton1,
Nicholas J Gay2 & Alicia Hidalgo1

Neurotrophin receptors corresponding to vertebrate Trk, p75NTR or Sortilin have not been identified in Drosophila, thus it is unknown
how neurotrophism may be implemented in insects. Two Drosophila neurotrophins, DNT1 and DNT2, have nervous system functions,
but their receptors are unknown. The Toll receptor superfamily has ancient evolutionary origins and a universal function in innate
immunity. Here we show that Toll paralogs unrelated to the mammalian neurotrophin receptors function as neurotrophin receptors
© 2013 Nature America, Inc. All rights reserved.

in fruit flies. Toll-6 and Toll-7 are expressed in the CNS throughout development and regulate locomotion, motor axon targeting and
neuronal survival. DNT1 (also known as NT1 and spz2) and DNT2 (also known as NT2 and spz5) interact genetically with Toll-6
and Toll-7, and DNT1 and DNT2 bind to Toll-6 and Toll-7 promiscuously and are distributed in vivo in domains complementary to or
overlapping with those of Toll-6 and Toll-7. We conclude that in fruit flies, Tolls are not only involved in development and immunity
but also in neurotrophism, revealing an unforeseen relationship between the neurotrophin and Toll protein families.

The Toll receptor superfamily, comprising Toll and Toll-like recep- proteases Easter, acting in development, and Spätzle Processing Enzyme
tors (TLRs), has ancient evolutionary origins, arising over 700 mil- (SPE), acting in immunity, to release the active cystine knot13. This
lion years ago, and is present throughout metazoans1. Toll and TLRs mechanism resembles the extracellular cleavage of BDNF at the synaptic
have a universal function in innate immunity, and they initiate adap- cleft by the serine protease plasmin (which is also involved in the blood-
tive responses in vertebrates1,2. In humans the ten TLRs are pattern clotting cascade) and which is activated by the presynaptic release of
recognition receptors that directly bind to microbial antigens and plasminogen activating factor (tPA) upon high frequency stimulation16.
activate proinflammatory and co-stimulatory responses. Mammalian The characteristic neurotrophin cystine knot, formed by antiparallel
TLRs were identified by homology to Drosophila Toll (Toll-1). The β-sheets held together by three intersecting disulfide bonds, can be pre-
Drosophila genome contains nine Toll receptor genes (Toll-1 to Toll-9), cisely aligned between the crystal structures of Spz and NGF17–19.
which, except for Toll-9, are phylogenetically distinct from the ver- DNT1 was identified independently as related to BDNF,
tebrate TLRs1. Thus, Drosophila Toll-1 to Toll-8 form one clade and using vertebrate neurotrophin sequences as query to search the
Toll-9 together with vertebrate TLRs form another3. Toll-1 func- Drosophila sequenced genome with bioinformatics tools20. DNT1
npg

tions in developmental processes, including the establishment of the was found to be spz2, a paralog of spz (refs. 20,21). Structural pre-
embryonic dorso-ventral axis, in axon targeting and degeneration, diction analysis showed that, of the spz paralogs, DNT1 and DNT2
and in innate immunity1,4, but the roles of the remaining Tolls are (spz5)21 are closest to the neurotrophin superfamily, followed by
largely unresolved. Reports have indicated that Toll-7 to Toll-9 have spz (refs. 20,21).
developmental functions but no antibacterial immunity functions, There is also functional conservation between DNT1, DNT2 and
although Toll-7 is involved in antiviral responses5–9 and Toll-6 and Spz and the mammalian neurotrophins in the nervous system20. The
Toll-7 are expressed in the CNS10. Unlike the TLRs, Toll-1 does not vertebrate neurotrophins have essential functions during develop-
bind microbial products directly. Instead, detection of bacterial mol- ment in neuronal survival, axon targeting and connectivity and during
ecules by the soluble recognition proteins PGRP and GNBP triggers a adult life in learning, memory and cognition16. During development,
serine protease cascade11. This leads to the cleavage and activation of DNT1, DNT2 and spz are expressed in target cells for CNS neurons,
Spätzle (Spz), an endogenous protein ligand for Toll-1 (ref. 12). such as the embryonic en-passant midline target of interneurons and
Spz belongs to the neurotrophin family of growth factors, which in the muscles, the target of motor neurons20. DNT1 and DNT2 are
vertebrates comprises nerve growth factor (NGF), brain-derived neu- required for neuronal survival, as neuronal apoptosis decreases upon
rotrophic factor (BDNF), neurotrophin-3 (NT3) and neurotrophin-4 their overexpression in the CNS and increases in the loss-of-function
(NT4) (refs. 13,14). Spz comprises a signal peptide, an unstructured mutants, leading to neuronal loss, with apoptotic neurons compris-
pro-domain and an active cystine knot domain of 13 kDa (also known ing those identified as bearing the Even-skipped (Eve) or Homeobox
C-106), which dimerizes, binding Toll with 2:2 stoichiometry13–15. Spz is 9 (HB9) neuronal markers20. DNTs are required for motor-axon
secreted as a pro-protein and is cleaved extracellularly by the serine targeting, as interfering with the function of DNT1, DNT2 and

1School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK. 2Department of Biochemistry, University of Cambridge, Cambridge, UK. 3These authors
contributed equally to this work. Correspondence should be addressed to A.H. (a.hidalgo@bham.ac.uk).

Received 15 February; accepted 20 June; published online 28 July 2013; doi:10.1038/nn.3474

1248 VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 nature NEUROSCIENCE


a r t ic l e s

Figure 1 Toll-6 and Toll-7 are expressed in the a Toll-6 mRNA b Toll-7 mRNA
CNS through all stages. In situ hybridizations
showing transcripts for Toll-6 and Toll-7 in (a,b)
stage 13, 15 and 17 embryos; (e,g) larval
optic lobes, central brain and ventral nerve
cords; and (i,k) adult brain in central complex 13 15 17 13 15 17
(arrows). (c,d,f,h,j,l) Distribution of GFP
immunoreactivity in Toll-6MIMICGFP and Toll-7
c GFP in Toll-6
MIMICGFP
d Toll-7
aCC U/CQ
immunoreactivity. (c) Arrows, GFP signal in ELs
distinct neuronal types. (d) White arrows in
first and second images from left, motor axons
exiting the CNS; yellow arrowheads, motor
17 16 17 13 14 14 14 17
neuron cell bodies; white arrows in third image,
axons crossing the midline; white arrows in e Toll-6 mRNA f Toll-6
MIMICGFP
g Toll-7 mRNA h Toll-7
fifth image, axons along three interneuron
fascicles and in thickenings (yellow arrowhead)
that might correspond to dendrites or glia.
(i,k) Arrows, signal in and around fan-shaped
body. ELs, U/CQ: Eve+ neurons. Anterior is up.
Scale bars: a,c, 10 µm; f,h,j,l, 50 µm.

Spz causes ­ misrouting, mistargeting and i Toll-6 mRNA j Toll-6MIMICGFP k Toll-7 mRNA l Toll-7
sprouting defects in motor axon terminals20.
Thus, DNT1 and DNT2, as well as Spz, are
© 2013 Nature America, Inc. All rights reserved.

Drosophila neurotrophins on the basis of


sequence, structural and functional homol-
ogy to the vertebrate neurotrophins20.
There is further cellular and molecular evi-
dence that neurotrophism operates in the Drosophila nervous system. death in several contexts25. Identified Drosophila neurotrophic factors
During normal Drosophila development, many neurons and glial cells include the homolog of mesencephalic astrocyte-derived neurotrophic
die22–24, and ablation or mutation in glial cells results in neuronal factor (MANF), which promotes dopaminergic neuron survival in
fruit flies using a noncanonical pathway26,
and Netrin, which promotes interneuron
a Merge MN Toll-6MIMICGFP b Merge MN Toll-7
survival from the en-passant midline tar-
get27. Gliotrophic factors of the transform-
ing growth factor (TGF)-α, neuregulin and
HB9

HB9

PVF/platelet-derived growth factor (PGDF)


protein families have also been shown to
c maintain glial survival in Drosophila22,28–30.
U/CQ
d
ELs
Lim3>RFP

Figure 2 Identification of Toll-6 and Toll-7 cells


Eve
npg

in the locomotor circuit. (a,c,e) Anti-GFP


RPs
in Toll-6MIMICGFP embryos is distributed in
ventral lateral nerve cord HB9+ neurons (a),
e f Eve+ EL interneurons and all Eve+ motor
IN
neurons (MN) except RP2 (c,e, arrows; for
RP2 neurons, see Supplementary Fig. 2c).
Fasll
Eve

IN, interneurons; arrowheads point to


longitudinal connectives. (b,d,f) Toll-7 protein
is localized to ventral lateral and medial HB9 +
g Toll-6>Tom Lim3>RFP Toll-7>GFP h Toll-6
MIMICGFP
Toll-6
MIMICGFP
Toll-7 neurons (b, arrows), Lim3GAL4UASmyrRFP
(Lim3>RFP) RFP+ RP motor neurons (d, arrows)
and possibly dendrites (pink arrow), and
to FasII+ interneuron fascicles (f, arrows).
(g) tdTomato in D42GAL4UAS10xmyr-tdTomato
(Toll-6>Tom) and GFP in w;Toll7GAL4/+;UASGAPGFP/+
(Toll-7>GFP) reveal ISNb/d terminals.
(h) Toll-6MIMICGFP and Toll-7 are present in the
i Merge TH Toll-6MIMICGFP j Toll-6
MIMICGFP
Toll-7 larval VNC neuropil (arrows point to axons)
and at least Toll-6MIMICGFP in motor neurons.
(i) Toll-6MIMICGFP colocalizes with the dopamine
precursor tyrosine hydroxylase (TH) in most
fsb dopaminergic neurons. (j) Toll-6MIMICGFP and
Toll-7 are distributed in distinct layers of the
fan-shaped body (fsb; arrows) and rings of the
ellipsoid body (eb; arrows). Scale bars: a–g,
eb 10 µm, h–j, 50 µm.

nature NEUROSCIENCE VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 1249


a r t ic l e s

Figure 3 Toll-6 and Toll-7 are required for a


larval locomotion and motor-axon targeting.
(a) Trajectories of larvae crawling for 400 frames
per larva, n = 50 larvae per genotype. (b) Toll-7;
Toll-6 double mutant larvae crawled more
slowly. Kruskal-Wallis = 814, P < 0.0001,
and Dunn’s test for pair-wise comparisons;
yw w; Toll-626/Toll-631 w; Toll-7P8/Toll-7P114 w; Toll-7P8/P114; Toll-626/31
asterisks refer to double mutants versus
yw controls (Dunn = 4,474), n = 50 larvae b 12.0 ***
and 19,950 frames per genotype. (c–e) The 10.0 yw

Percentage frames
incidences of FasII+ motor axon misrouting in 26 31
8.0 w; Toll-6 /Toll-6
one or more projections and of loss of two or
more projections per hemisegment increased 6.0 w; Toll-7P8/Toll-7P114
in stage 17 mutant embryos (c,e) and embryos 4.0 w; Toll-7P8/Toll-7P114; Toll-626/Toll-631
overexpressing activated forms of Toll-6 or Toll-7 2.0
in all neurons (elavGAL4>Toll-6CY;Toll-6CY and
0
Toll-7GAL4;elavGAL4>Toll-7CY) (d,e). Scale bar,

0. 8
0. 6
0. 5
0. 3
0. 1
0. 9
0. 7
0. 6
0. 4
0. 2
1. 0
1. 7
1. 5
1. 3
1. 1
1. 9
1. 8
1. 6
1. 4
1. 2
1. 0
1. 9
2. 7
05
10 µm. For e, chi-squared χ2(7) = 136.247,

0
1
2
3
4
4
5
6
7
8
9
0
1
2
3
3
4
5
6
7
8
8
9
0.
P < 0.001, pair-wise comparisons to yw Speed (mm s–1)
chi-squared with Bonferroni correction, c
n = 169–465 hemisegments per genotype. 12
**P < 0.01; ***P < 0.001. For details, 13
6
see Supplementary Table 1.
7
© 2013 Nature America, Inc. All rights reserved.

The mammalian neurotrophins signal


through three distinct receptor types—
p75NTR, Trk and Sortilin—and share a down-
stream target, the activation of NF-κB31–33. In Toll-7P114/ Toll-7P8/P114;
Toll-626/31 Toll-631/Df(3L)XG4 Toll-7
yw P8/P114
Drosophila there are no canonical homologs of Df(2R)BSC22 Toll-626/31

these receptors. The receptors for DNT1 and d Neurons GAL4 e 80


Fan
DNT2 are unknown, although one hypoth- 70 *** ***
Percentage hemisegments ≤1 loss
esis is that orphan Tolls fulfill this function in 60 ***
*** *** >1 loss
insects. Toll receptors are generally thought 50
to function by activating NF-κB signaling, 40 ** Misrouting

which regulates the production of antimicro- 30

bial peptides in immunity1. Neurotrophins 20


34
also function in immunity , but these roles 10
0
have been largely unexplored. TLRs are also UASToll-6CY UASToll-7CY
31

14

1
yw

Y
/3
present in the CNS, primarily in microglia,
4

22

C
P1

26
XG
l-6

7
SC

ll-

ll-
l-7

l-6
ol

L)

To

To
where they have immunity-related functions35. Thus, potential rela-
)B
/T

f(3

ol

ol
26

4>

4>
R
/T

;T
P8
/D

f(2

4
6
31

AL

AL
11
ll-

tionships between the Toll and neurotrophin families may have been
7

/D
/P
To

G
P8
ll-

P8
ll-

ns

ns
To
To

ro

ro

overlooked. Here we ask whether Toll-6 and Toll-7 can function as


7
npg

ll-

ll-
eu

eu
To

To

receptors for DNT1 and DNT2 during CNS development.


both interneurons and motor neurons (Fig. 1c,d), but we cannot rule
RESULTS out expression also in glia. Toll-6MIMICGFP is present in ventral embry-
Toll-6 and Toll-7 are expressed and required in the CNS onic HB9+ neurons (Fig. 2a) in all Eve+ motor neurons except raw
Toll-6 and Toll-7 mRNA are expressed in the embryonic and larval prawn 2 (RP2) (Fig. 1c, Fig. 2 and Supplementary Fig. 2c) and in
CNS and adult central brain (Fig. 1). To visualize protein distribu- longitudinal interneuron axons (Fig. 2e). Toll-7 protein is distributed
tion, we used a GFP exon trap insertion into Toll-6 (hereafter named in ventral HB9+ and LIM-homeodomain-3 (Lim3)+ RP motor neu-
Toll-6 MIMICGFP), and we raised antibodies to Toll-7. The rons (Fig. 1d, Fig. 2b,d and Supplementary Fig. 2d), in interneuron
Toll-6MIMICGFP insertion is likely to result in a truncated Toll-6 protein axons that cross the midline (Fig. 1d) and project along the three
and thus a mutant allele that could conceivably affect expression from fasciclin II (FasII)+ longitudinal fascicles (Figs. 1d and 2f), and pos-
the locus. However, we have no evidence that Toll-6 regulates its own sibly in motor neuron dendrites or perhaps glia (Fig. 2d). Ventral
expression. The Toll-7 antibodies were found to be specific, as they HB9+ and Lim3+ motor neurons project along the intersegmental
did not detect signal in Toll-7 null mutant embryos or in deficiency nerve b/d (ISNb/d), which targets muscles 6,7,12,13. The above dis-
embryos lacking the Toll-7 locus (Supplementary Fig. 1a). The distri- tributions overlap with those of GAL4 reporters for Toll-6 (ref. 36)
bution of GFP in Toll-6MIMICGFP and of Toll-7 in wild-type matched and Toll-7 (Supplementary Fig. 2a,b,e–i), which include expression
the expression patterns of Toll-6 and Toll-7 transcripts, respectively in the ISNb/d axonal terminals (Fig. 2g). In the larva, immunore-
(compare Fig. 1a,b,e,g,i,k with Fig. 1c,d,f,h,j,l). This indicates that activity for GFP in the Toll-6MIMICGFP and immunoreactivity for
the protein patterns most likely represent the endogenous distribu- Toll-7 were distributed along the ventral nerve cord (VNC) neuropil
tion of the receptors. In Toll-6 MIMICGFP, GFP is mostly cytoplasmic, (Figs. 1f,h and 2h), and Toll-6MIMICGFP is detectable in anterior cor-
whereas Toll-6 is localized to the membrane; therefore, the GFP does ner cell (aCC) motor neurons (Fig. 2h and Supplementary Fig. 2i).
not reveal the subcellular distribution of Toll-6. Toll-6MIMICGFP and In the adult brain, Toll-6MIMICGFP is distributed in dopaminergic neu-
Toll-7 proteins are distributed in the CNS (Fig. 1c,d,f,h,j,l), including rons (Fig. 2i). Toll-6MIMICGFP and anti–Toll-7 immunoreactivity are

1250 VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 nature NEUROSCIENCE


a r t ic l e s

Figure 4 Toll-6 and Toll-7 maintain neuronal a b


** *
c *** *
d * *
survival. (a) Embryonic VNCs labeled with 200 180
Caspase 175
anti–cleaved-Caspase-3. (b,c) Apoptosis 180 160

No. Caspase+ cells


increased in Toll-7 and Toll-6 mutant embryos, 150 140
160
as quantified with DeadEasy software: 125 120
140
one-way ANOVA F(2,70) = 5.782, P = 0.005; 100
100 120
post hoc Dunnett P = 0.006, P = 0.015, 80
respectively; n = 19–28 embryos per genotype 100
75 60
(b); one-way ANOVA F(2,71) = 7.010, 80

P = 0.002; post hoc Dunnett P = 0.001, P8 P114 yw


26
Toll-6 /Toll-6
31 26
Toll-6 /Df(3L)XG4 ElavG4>Toll-6
CY
yw Toll-7 /Toll-7 P8 P114 P8 CY
P = 0.032, respectively; n = 21–31 Toll-7 /Toll-7 Toll-7 /Df(2R)BSC22 ElavG4>Toll-7
embryos (c). (d) Pan-neuronal overexpression e f g h yw
Toll-7P8/Toll-7P114;
of activated Toll-6 and Toll-7 rescued
Toll-626/Toll-631
naturally occurring cell death in the CNS;

Merge

Merge

Merge
24
*
one-way ANOVA F(2,68) = 4.811, P = 0.011; 22
20

Caspase HB9 cells


post hoc Dunnett P = 0.021, P = 0.012, 18
respectively; n = 22–27 embryos. In b–d, 16

+
HB9

HB9
HB9
14
asterisks refer to pair-wise comparisons to 12

+
10
yw, post hoc Dunnett tests. (e–h) Apoptotic 8

Caspase
Caspase

Caspase
Caspase+HB9+ cells in Toll-7P8/Toll-7P114; 6
4
Toll-626/Toll-631 double mutant embryos 2
0
in locations corresponding to neurons that –/–
P8
Toll-7 /Toll-7
P114 26
; Toll-6 /Toll-6
31 P8
Toll-7 /Toll-7
P114 26
; Toll-6 /Toll-6
31
yw Toll-7 –/–;
normally express Toll-6 (e) or Toll-7 (f), high Toll-6
magnification view (g) and quantification (h); i j k l
© 2013 Nature America, Inc. All rights reserved.

yw ELs Us/CQs ELs ELs Eve ELs yw


unpaired Student t-test t(1) = −2.230, Toll-7P8/Toll-7P114; Toll-7P8/Toll-7P114;
Merge

P = 0.035, n = 9–19 embryos. (i,j) In 26


Toll-6 /Toll-6 31
Toll-626/Toll-631
Toll-7P8/Toll-7P114; Toll-626/Toll-631 20 25
Percentage EL clusters

**
with Cas+ Eve+ cells

18

clusters with ≤7 cells


Percentage Eve+ EL
double mutant embryos, more (albeit
Eve EL

16 20
14
not significantly more) EL clusters had 12 15
Eve+Caspase+ apoptotic interneurons compared 10
8 10
Caspase

to wild-type (yw) controls (j, χ (1) = 1.992,


2 6
4 5
P = 0.158, n = 109–138 EL clusters). 2
+ 0 0
(k,l) Apoptosis leads to loss of Eve EL
yw Toll-7P8/Toll-7P114; Toll-626/Toll-631
interneurons in the double mutants. More EL
clusters had fewer neurons than the normal 8–10 per cluster (arrowheads in k); chi-squared χ2(1) = 9.645, P = 0.002, n = 22–260 EL clusters. *P < 0.05;
**P < 0.01; ***P < 0.001. All embryos stage 17. Scale bars: a, 20 µm; e,f,k, 10 µm; g,i, 5 µm. For details, see Supplementary Table 1.

distributed in complementary layers of the fan-shaped body and in and P = 0.032, respectively). Overexpression of Toll-6CY and Toll-7CY
complementary rings of the ellipsoid body (Fig. 2j), the sites for the in neurons rescued naturally occurring cell death (Fig. 4d, ANOVA,
central control of locomotion. Thus, Toll-6 and Toll-7 are distributed F(2,68) = 4.811, P = 0.011; corrected P = 0.021, P = 0.012, respec-
in the locomotor circuit, including motor neurons, interneurons of tively). Thus, Toll-6 and Toll-7 can promote cell survival. The increase
the central pattern generator, and locomotion centers and dopamin- in dying cells in the double mutants compared to wild-type controls
ergic neurons in the brain. affected HB9+Caspase+ neurons (Fig. 4e–h, Student t(26) = −2.230,
To investigate the functions of Toll-6 and Toll-7 in the CNS, we P = 0.035) and, albeit not significantly, Eve+Caspase+ EL interneurons
npg

generated null mutant alleles (Supplementary Fig. 3). The embryonic (Fig. 4i,j, χ2(1) = 1.992, P = 0.158), which normally express Toll-7 and
motor neurons are preserved in the larva; thus, we tracked crawling Toll-6 (Fig. 2a–f). Not all HB9+ neurons normally express Toll-6 or
mutant larvae. Most particularly, Toll-7P8/Toll-7P114;Toll-626/Toll-631 Toll-7, and thus we could not confirm that all dying HB9+ neurons
double mutants crawled more slowly than controls (Fig. 3a,b, necessarily corresponded to Toll-6+ or Toll-7+ neurons. However,
P < 0.0001, corrected P < 0.001). To test whether Toll-6 and Toll-7 there was a good correlation between the ventral and central locations
function in motor-axon targeting, we visualized the projections of the dying HB9+ neurons in the double mutants and the equivalent
of the FasII + ISNb/d. Toll-631/Df(3L)XG4, Toll-7P8/Toll-7P114 location of HB9+ Toll-6MIMICGFP-positive and HB9+Toll-7+ neurons
and Toll-7114/Df(2R)BSC22 single mutants and Toll-7P8/Toll-7P114; in normal embryos (compare Fig. 4e with Fig. 2a, and Fig. 4f with
Toll-626/Toll-631 double mutants showed deficient targeting and Fig. 2b), indicating that, in the mutants, the HB9+ dying neurons most
axonal misrouting (Fig. 3, χ2(7) = 136.247, P < 0.001, corrected likely included Toll-6+ and Toll-7+ neurons. We were able to confirm
P < 0.001). Overexpression of constitutively active forms of the that because all Eve+ neurons except RP2 were also Toll-6MIMICGFP
receptors, Toll-6CY and Toll-7CY, in neurons also caused targeting positive, the apoptosis of Eve+ neurons in the mutants corresponded
defects (Fig. 3d,e, χ2(7) = 136.247, P < 0.001; corrected P = 0.032 and to cell death of at least Toll-6+ neurons. Apoptosis resulted in neu-
P < 0.001, respectively). To test whether Toll-6 and Toll-7 can regulate ronal loss, as in Toll-7P8/Toll-7P114;Toll-626/Toll-631 doubles mutants
cell survival, we visualized cell death with anti–cleaved-Caspase-3 there was a reduction in the number of Eve+ EL interneurons
antibodies and quantified the number of apoptotic cells using (Fig. 4k,l, χ2(1) = 9.645, P = 0.002). Altogether, our data show that
DeadEasy Caspase software37. Apoptosis increased in Toll-626/Toll-631, Toll-6 and Toll-7 are required for locomotion, motor-axon targeting
Toll-626/Df(3L)XG4, Toll-7P8/Toll-7P114 and Toll-7P8/Df(2R)BSC22 and neuronal survival.
mutant embryos compared to that in wild-type controls, showing that
Toll-6 and Toll-7 are required for cell survival in the CNS (Fig. 4a–c) Toll-6 and Toll-7 interact genetically with DNT2 and DNT1
(Fig. 4b: F(2,70) = 5.782, P = 0.005; corrected P = 0.006 and P = 0.015, The observed CNS phenotypes resemble those caused by DNT1 and
respectively; Fig. 4c: F(2,71) = 7.010, P = 0.002; corrected, P = 0.001 DNT2 (ref. 20), which is consistent with DNTs and Tolls being involved

nature NEUROSCIENCE VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 1251


a r t ic l e s

a Single Double Single mutants Double b Controls ChaGAL4


1.2 0.5 ***
1.0 0.4

Survival index
Survival index

0.8
0.3 *** **

R
R

R
∆L

Y
∆L
0.6

C
6

7
0.2

ll-
4

ll-

ll-
al

To

To

To
0.4

AS
*** *** *** *** ***

AS

AS
ha
0.1

U
C

U
0.2
0 0
UAS: Toll-6∆LRR Toll-7
∆LRR
Toll-7CY
T

T1

T2

T1

T1

l-6

l-6

l-6
ll-

ll-

ll-

ll-
W

ol

ol

ol
To

To

To

To
All samples are Toll-7P8;Toll-626 double mutant
D

7T

7T

7T
T2

T2

ll-

ll-

ll-
N

To

To

To
D

c 1.4
Toll-7GAL4;
DNT1 Toll-6 d Controls ElavGAL4
1.0
1.2 *** ***
0.8 *** ***

R
R

R
1.0
Survival index

Survival index

Y
∆L

∆L

b
10
6

7
4
0.8 0.6 *** ***

ll-

ll-

ll-

ll-
AL

ll
To

To

To

To

To
G
***

AS

AS

AS

AS

AS
av
0.6 0.4

El

U
0.4
0.2
0.2
0 0
DNT1 DNT2 Toll-7 Toll-7

b
Y

Y
CY
UASToll-7CY UAS:

10
C

C
R

R
UASToll-6

∆L

∆L
Toll-6 Toll-6 DNT1 DNT2

ll
ll-

ll-

To
6

To

To
ll-

ll-
To

To
e f All samples are DNT2e03444DNT141 double mutant
© 2013 Nature America, Inc. All rights reserved.

175 * 130
* * g
No. Caspase cells

No. Caspase cells

150 150
Controls ElavGAL4
0.7
125 125
0.6 ***
Survival index

100 100 0.5


* ***

Y
0.4

C
b
10
75 75

7
4

ll-

ll-
AL

ll
0.3

To

To
To
G
2

AS

AS
AS
av
z
sp

0.2
El

U
U
55 CY
yw control DNT1 , ElavGAL4>Toll-7 0.1
DNT155 DNT2e03444, ElavGAL4>Toll-6CY 0
DNT2e03444/Df6092 All samples are spz2 mutant UAS: Toll10b Toll-6CY Toll-7CY

Figure 5 Toll-6 and Toll-7 interact genetically with DNT2 and DNT1. Survival index for homozygous yw;;+/+ controls bred from an outcross to TM6B
at 18 °C is 1. (a) Single homozygous mutants lacking one DNT or Toll-6 or Toll-7 are viable, whereas homozygous double losses of DNT1 and DNT2 or
Toll-6 and Toll-7 are semi-lethal if bred at 18 °C as progeny of a stock maintained over a TM6B or SM6aTM6B balancer. Chi-squared χ2(11) = 360.277,
P < 0.001, n = 126–872 pupae per genotype. (b) The semi-lethality of Toll-7P8;Toll-626 double mutation can be rescued by overexpressing the activated
receptors in cholinergic neurons. χ2(6) = 85.028, P < 0.001, n = 102–467 pupae. (c) Homozygous double losses of one DNT and one Toll recapitulate the
semi-lethality of DNT141DNT2e03444 and Toll-7P8;Toll-626 double mutations, and the lethality DNT1 Toll-6 double mutations can be rescued by expressing
the activated receptors with Toll-7GAL4. χ2(5) = 653.525, P < 0.001, n = 72–991. (d) The semi-lethality of DNT141DNT2e03444 double mutation can be
rescued by expressing activated Toll-6, Toll-7 or Toll-1 receptors in neurons. χ2(10) = 401.419, P < 0.001, n = 83–1,461 pupae. (e,f) Quantification of
npg

anti–cleaved-Caspase-3 labeling in embryonic VNCs: apoptosis increase in DNT55 and DNT2e03444/Df6092 mutant embryos compared to wild-type (yw)
controls (e, one-way ANOVA F(2,69) = 10.479, P < 0.001; post hoc Dunnett P < 0.01, P = 0.051, respectively) is rescued with the overexpression of
activated Toll-7CY and Toll-6CY in all neurons (f, Welch ANOVA F(2,63) = 5.143, P = 0.009; post hoc Dunnett P = 0.011, P = 0.017, respectively). In e,f,
asterisks refer to pair-wise comparisons to yw, post hoc Dunnett tests. (g) Pan-neuronal overexpression of activated Toll10b, Toll-6CY and Toll-7CY rescues the
semi-lethality of the spz2 mutation; χ2(7) = 99.272, P < 0.001. ***P < 0.001; **P < 0.01; *P < 0.05. (a–d,g) Asterisks refer to chi-squared comparisons
to fixed controls with Bonferroni corrections. For detailed genotypes and statistics details, see Supplementary Tables 1 and 2.

in common developmental processes. We next asked whether DNT, embryos are viable (Fig. 5c). The semi-lethality of DNT141Toll-626
Toll-6 and Toll-7 mutants might interact genetically. Single DNT1, and Toll-7P114;DNT2e03444 double mutation is consistent with lack of
DNT2, Toll-6 or Toll-7 mutants are viable. However, DNT1 DNT2 the receptor from one signaling pathway and the ligand from the other
double mutation is semi-lethal in progeny of a heterozygous stock being equivalent to losing both ligands or both receptors. The viability
maintained over the TM6B chromosome at 18 °C (Fig. 5a, χ2(11) = of DNT2e03444Toll-626 double mutants is consistent with lack of both
360.277, P &lt; 0.001, corrected P < 0.001). Similarly, Toll-7;Toll-6 the receptor and the ligand from the same pathway being equivalent
double mutation is also embryonic semi-lethal at 18 °C in progeny of to but not worse than losing only one of them. The semi-lethality of
a heterozygous stock over SM6aTM6B (Fig. 5a, corrected P < 0.001). DNT141Toll-626 double mutation can be rescued by the overexpres-
This semi-lethality can be rescued with the expression of consti- sion of Toll-6CY or Toll-7CY with Toll-7GAL4 (Fig. 5c, χ2(5) = 653.525,
tutively active forms of the receptors in cholinergic interneurons, P < 0.001; corrected P < 0.001, P < 0.001 respectively). These data
Toll-6∆LRR, Toll-7∆LRR and Toll-7CY (see Online Methods) (Fig. 5b, suggest that Toll-7 may function downstream of DNT1 and Toll-6
χ2(6) = 85.028, P < 0.001; corrected P < 0.001, P = 0.003, P < 0.001, downstream of DNT2. DNT141Toll-7P114 double mutants also have
respectively). Exploiting this cold-sensitive semi-lethality, we tested reduced viability, suggesting that Toll-7 may also act downstream
genetic interactions between the DNTs and the Tolls. DNT141Toll-626 of DNT2.
and Toll-7P114;DNT2e03444 double mutation is semi-lethal under We thus asked whether activated forms of Toll-6 and Toll-7 could
the above conditions, whereas DNT2e03444Toll-626 double mutant rescue DNT mutant CNS phenotypes. Pan-neuronal expression of

1252 VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 nature NEUROSCIENCE


a r t ic l e s

Figure 6 In vitro, cell culture and in vivo

g
a b

in
ECD

c
To ECD

D
evidence that Toll-7 and Toll-6 bind DNT1 and

on ing
du
C
7E

-re
uc
6
ll-

ll-
DNT2. (a) The constructs encoding tagged

ed
R

CR
DNT1 DNT2

To
R
R

R
TM

A
SP

LR

LR

R
N
TI

IT
C

H
Full length
proteins. (b) Coomassie stainings showing Toll-6, Toll-7 250 50 58
37 46
secreted Toll-6ECD and Toll-7ECD purified from

AG
130 25 CK CK,

CR

F L is
30

R
SP

LR

LR

6H
dimer CTD

C
20 25
S2-cell conditioned medium (left panel; for Toll-6ECD, 100
Toll-7ECD 15 CK
mass spectrometry, see Supplementary Fig. 5e); 70
10
17
CK

TD
V 5 is
6HV
o

K
SP

TE
Anti-V5
DNT2 purified from a baculovirus expression

Pr

C
DNT1
system as a secreted cleaved cystine knot (CK) S2 cells
DNT1
c A
26,225.5

(% ion mass/charge)
Mass = 13,113.65 Da
dimer (middle panel); and DNT1 purified in vivo 1–116

DNT2 Gln1 + –17.03


from S2 conditioned medium as cleaved

Intensity
S2 cells 1 25

45-kDa cystine knot plus terminal extension DNT2


Baculovirus 26,246.0
26
51
50
75

(CK, CTD; left arrow), plus full-length form and 26,202.0


26,182.0
26,269.0
76
101
100
116

cleavage products, and DNT2 purified from


S2 conditioned medium as cleaved CK (right
d – DNT2His + DNT2His 2 Mass

00

00

00

00

00

00

00
To -7E DH His

is

,0

,4

,8

,2

,6

,0

,4
H

25

25

25

26

26

27

27
ll C is
l 7 is

ll- CD is
7E is
ll C D

D
arrow) only (right panel). (c) The mass of DNT2

To l-7E DH
To -6+ DH

6+ H
e

To l-6E EC

C
ll is
l C
Toll-7ECD-6His-3xFLAG: MW/pl = 121,578.21/6.00

To T2H
purified by reverse phase chromatography,

To -6E
Toll-6ECD-6His-3xFLAG: MW/pl = 124,778.08/6.06

N
D
as determined by MALDI TOF mass Cathode (–) 150 DNT2CK-TEV-6His: MW/pl = 13,112.65/7.94
100
spectrometry. The N-terminal residue and the

Net charge
50 Buffer pH
seven cysteines that form three intra- and one 0
Anode (+) 3 4 5 6 7 8 9 10
inter-molecular disulfide bond are highlighted. WB: His
–50
–100
Gln1 + −17.03 indicates that approximately –150 pH
17 Da are lost due to cyclization of the N-terminal Key for ELISA 1. No DNA 2. Toll-6HA 4. DNT1V5 6. Toll-6HA + DNT2V5
glutamine to pyroglutamate. Together with 7 Da and co-IP (f–i): 3. Toll-7HA 7. Toll-7HA + DNT1V5
5. DNT2V5
© 2013 Nature America, Inc. All rights reserved.

eliminated in the formation of seven disulfide


bonds, this gives an expected mass for the f
Control 1 Control 2 Control 3 Control 4
ELISA
Capture V5 Detection HA g
DNT2 monomer of 13,112.65 Da, or a dimer 1 2 3 6 7 1 2 3 6 7 1 4 5 6 7 1 4 5 6 7 *** **
2.5
mass of 26,225.3 Da. The observed mass of 250 T6/7 DNT1

Absorbance fold
100 FL FL 2.0
peak A is 26,225.5 Da. (d) Native gel showing

difference
55 DNT1
complexes of purified DNT2CK with purified 35 DNT2 CK
1.5

Toll-6ECD, Toll-7ECD or both Toll-6ECD plus 25 FL 1.0


Toll-7ECD. WB, western blot. (e) Predicted 0.5
15 DNT2
mobility of native Toll-6ECDHisFLAG, Toll- CK 2 6 3 7
7ECDHisFLAG and DNT2CKHis at pH 8.8. S2 lysate WB: HA S2 lysate WB: V5 S2 lysate WB: V5 S2 lysate WB: HA Capture HA Detection V5
(f) Cotransfection S2 cell lysate controls for
ELISA and coimmunoprecipitation (co-IP)
h
Control
1 4 5 6 7
S2 cell co-IP
1.5
** ***
DNT1–Toll-7 DNT2–Toll-6
experiments showing proteins expressed in 1.4

Absorbance fold
250 Toll-6/7 1 4 7 1 5 6

difference
DNT1 FL 1.3
each experiment in g–i. (g) ELISAs using 100
FL
DNT1 55 1.2
cotransfected S2 cells revealed a significant 70 55 DNT2 FL 1.1
difference in absorbance comparing singly 55 1.0
IP: HA WB: V5 IP: HA WB: V5
transfected and cotransfected S2 cell lysates. 35 0.9
Unpaired t-tests: top, condition 6 versus 2: 4 7 5 6
t(4) = −10.485, P < 0.001; 7 versus 3: IP: HA WB: HA Key for in vivo co-IP: 8. Wild type
t(4) = −7.619, P = 0.002; bottom, 7 versus 4: 9. GMRGAL4>Toll-7HA
i j + DNT1FLAG
npg

t(4) = −5.574, P = 0.005; 6 versus 5: Control S2 cell co-IP Control Control


t(4) = −13.504, P < 0.001; n = 3 repeats. 1 2 3 6 7
DNT2–Toll-6, DNT1–Toll-7
8 9
250
8 9 In vivo co-IP
250 Toll-7 8 9 8 9
(h,i) Co-IP of full-length Toll-7HA and DNT1V5, 100 DNT1 FL FL
Toll-7
1 2 3 6 7 130 FL
and full-length Toll-6HA and DNT2V5, from 55
250
DNT2FL 100
cotransfected S2 cells. (h) Precipitation of 35 DNT1 250 Toll-6/7
70
CK IP: FLAG WB: HA
receptors with anti-HA coprecipitates bound 25 CTD FL
130 55 DNT1
ligands detected with anti-V5. (i) Precipitation 15 IP: V5 WB: HA
of ligands with anti-V5 coprecipitates bound DNT2 CK Head Iysate Head Iysate
WB: HA WB: FLAG
receptors detected with anti-HA. (j) In vivo co-IP IP: V5 WB: V5
from transgenic flies overexpressing full-length
Toll-7HA and DNT1-FLAG in the retina with GMRGAL4. Two examples are shown, using rabbit (left) or mouse (right) anti-Flag antibodies to precipitate DNT1,
coprecipitating bound receptor detected with anti-HA. ***P < 0.001, **P < 0.01, *P < 0.05. (h–j) Full-length blots are shown in Supplementary Figures 6–9.

Toll-6∆LRR, Toll-7∆LRR, Toll-6CY or Toll-7CY rescued the semi-lethality Toll-6 and 7 function upstream of NF-kB and bind DNT1 and 2
of DNT141DNT2e03444 double mutation (Fig. 5d, χ2(10) = 401.419, We next asked whether potential interactions between the DNT1 and
P < 0.001, corrected P < 0.001 for all). Overexpression of Toll-7CY DNT2 ligands and Toll-6 and Toll-7 receptors could induce NF-κB
in all neurons rescued the apoptosis caused by loss of DNT1 func- signaling. Pan-neuronal overexpression constitutively active Toll10b,
tion, and overexpression of Toll-6CY in all neurons rescued the apop- which activates NF-κB homologs Dorsal and Dorsal-related immunity
tosis caused by loss of DNT2 function (Fig. 5e, F(2,69) = 10.479, factor (Dif)4, rescued the semi-lethality in spz2 mutants, and this res-
P < 0.001, and Fig. 5f, F(2,63) = 5.143, P = 0.009; corrected P < 0.01, cue was replicated with the overexpression of activated Toll-6CY and
P = 0.051, P = 0.011, P = 0.017, respectively). Altogether, these data Toll-7CY in neurons (Fig. 5g, χ2(7) = 99.272, P < 0.001; corrected
indicate that Toll-7 and Toll-6 most likely function as receptors for P < 0.001, P = 0.018, P < 0.001, respectively). Transfection of the S2
DNT1 and DNT2, respectively, although these interactions may cell line with activated Toll-6CY and Toll-7CY resulted in the activa-
be promiscuous. tion, upon induction, of snail-luciferase, a reporter for Dorsal, and

nature NEUROSCIENCE VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 1253


a r t ic l e s

Figure 7 The relative distributions of DNT1, 2 and Toll-7, 6, respectively, a DNT1 b DNT1 c DNT2 Fasll Fasll d DNT2 D42
in vivo are consistent with their functions are ligand-receptor pairs.
(a) Anti-DNT1 reveals DNT1 protein distributed in the embryonic CNS
midline (stage 15) and (b) at high levels in muscle 13,12, in lower
levels in muscles 6,7 and possibly others too (stage 17). (c,d) Anti-DNT2
reveals punctate signal along larval CNS axons revealed with (c) FasII+,
(d) DsRed+ in Toll-6GAL4(D42)>myrRFP and (e) Toll-6MIMICGFP. (f) Anti- Toll-6GFP DNT2
DNT1 and anti–Toll-7 colocalize in fan-shaped-body layers. Anti-DNT2 e
and anti-GFP in Toll-6MIMICGFP are distributed in complementary fan-
shaped-body layers. Anterior is up. Scale bars: a,d,e, 10 µm; c,f, 50 µm.
f Merge DNT1 Toll-7

drosomycin-luciferase, a reporter for Dif (Supplementary Fig. 4a,

DNT1 Toll7
F(5,24) = 27.165, P < 0.001; corrected P = 0.054, P = 0.000084, respec-
tively; Supplementary Fig. 4b, F(5,24) = 6.574, P = 0.001; corrected
P = 0.01, P = 0.018, respectively). Furthermore, when S2 cells trans-
fected with hemagglutinin (HA)-tagged Toll-6HA or Toll-7HA were Merge DNT2 Toll-6GFP

DNT2 Toll-6GFP
stimulated with purified DNT2, this triggered a drosomycin-luciferase
readout indicative of Dif signaling (Supplementary Fig. 4c,d,
F(5,27) = 16.788, P < 0.001, corrected P = 0.034, P = 0.09, respectively;
and Supplementary Fig. 5a–d). However, in vivo, overexpression
of Toll-6CY and Toll-7CY did not induce drosomycin-GFP expression
(Supplementary Fig. 4e), as is consistent with previous reports7,8. from cotransfected cells (Fig. 6h). There was some nonspecific bind-
© 2013 Nature America, Inc. All rights reserved.

Nevertheless, activated Toll-6CY and Toll-7CY induced increases in ing of DNT1 in the control lacking receptor, but at lower levels than in
Dorsal, Dif and Cactus proteins (Supplementary Fig. 4f; Dorsal: the cotransfected cells (Fig. 6h). In the reverse coimmunoprecipita-
F(2,9) = 10.382, P = 0.005; corrected P = 0.003, P = 0.085, respectively; tion, anti-V5 precipitated full-length and cleaved V5-tagged DNT2
Dif: F(2,9) = 12.898, P = 0.005; corrected P = 0.002, P = 0.006, respec- and DNT1, and did so only in ligand-transfected cells (Fig. 6i). By
tively; Cactus: F(2,4.14) = 28.233, P = 0.004; corrected P = 0.038, contrast, we detected no receptors after immunoprecipitation from
P = 0.011, respectively). Although we do not provide mechanistic control S2 cells that had not been transfected with ligands, whereas
evidence for whether or not Toll-6 or Toll-7 signaling involves the antibodies specific for the V5-tagged forms of DNT1 and DNT2 copu-
canonical Toll pathway, our data indicate that Toll-6 and Toll-7 func- rified Toll-7HA and Toll-6HA, respectively, from cotransfected cells
tion upstream of NF-κB. (Fig. 6i). Together, these data show that DNT1 or DNT2 and full-
In the light of the genetic evidence that Toll-6 and Toll-7 are recep- length transmembrane Toll-6 or Toll-7 can be coimmunoprecipitated
tors for Drosophila neurotrophins, we next asked whether DNT1 from S2 cells. In vivo, we immunoprecipitated DNT1 bound to Toll-7
and DNT2 could bind Toll-7 and Toll-6. To test whether they can from transgenic flies overexpressing (using a GMRGAL4 driver) both
interact in vitro, we purified His-tagged secreted forms of the recep- Flag epitope–tagged DNT1 cystine knot domain (DNT1-CK-Flag)
tors comprising only the extracellular domain, Toll-6-ECDHis and and full-length Toll-7HA in the retina (Fig. 6j). Together, these data
Toll-7-ECDHis (Fig. 6a,b and Supplementary Fig. 5e); and a demonstrate that DNT1 binds Toll-7 and DNT2 binds promiscuously
baculovirus-expressed cleaved DNT2 cystine knot domain, DNT2- Toll-6 and Toll-7.
CK-His (Fig. 6a–c and Supplementary Fig. 5a–d). We mixed these Consistent with their functions as ligands for Toll-7 and Toll-6 in
to allow formation of complexes, which were subjected to native gel interneurons and motor neurons, DNT1 and DNT2 are expressed
npg

electrophoresis (Fig. 6d,e). In native gels, protein mobility does not in embryonic CNS midline and muscle20. We found anti-DNT1
depend on molecular weight but on conformation and charge relative antibodies to be specific, as they did not reveal signal in DNT141
to the pH of the buffer. At the pH of our buffers, all three proteins were null mutant embryos (Supplementary Fig. 1b). In normal embryos,
negatively charged, but DNT2CK just barely so, as its pI was close to DNT1 protein was detectable at the midline (Fig. 7a), a target of
the buffer pH. Thus the mobility of the latter was limited, whereas inter­neurons, and in high levels in muscles 13,12 and lower levels in
the Toll-6ECD and Toll-7ECD migrated further (Fig. 6d), as their muscles 6,7, the targets of ISNb/d axons (Fig. 7b). We were not able
pI values differed from the buffer pH. Adding DNT2-CK shifted the to generate a DNT2 null allele; thus, we could not confirm that anti-
mobility of Toll-6ECD and Toll-7ECD, and new bands appeared at DNT2 signal was absent in mutants. However, anti-DNT2 detected
the top of gel, relative to those in controls (Fig. 6d,e). This indicates ectopic DNT2 distribution in embryos, larval and adult brains
that DNT2 interacts with both Toll-6 and Toll-7. (Supplementary Fig. 1c–e), suggesting that it detects endogenous
In S2 cell culture, cotransfection with full-length DNT1 tagged DNT2 protein in vivo. In normal larvae, anti-DNT2 revealed puncta
with the simian virus 5 (V5) epitope (DNT1V5) and full-length along FasII+ interneuron axons (Fig. 7c), Toll-6GAL4>myrRFP RFP+
Toll-7HA, and with DNT2-V5 and Toll-6HA (Fig. 6f), revealed (Fig. 7d) and Toll-6MIMICGFP GFP+ (Fig. 7e) axons. In adult brains,
interactions between DNT1 and Toll-7 and between DNT2 and DNT1 overlay Toll-7 in fan-shaped-body layers (Fig. 7f), whereas
Toll-6 in ELISAs (Fig. 6g). This interaction was also demonstrated DNT2 and Toll-6MIMICGFP were distributed in complementary layers
by coimmunoprecipitation of the ligands and receptors expressed in (Fig. 7f), compatible with the nonautonomous function of DNT2.
cotransfected S2 cells. Anti-hemagglutinin precipitated full-length Thus, the distribution of DNT1 and DNT2 supports their functioning
Toll-6HA and Toll-7HA receptors, and did so only in receptor- as Toll-7 and Toll-6 ligands in vivo.
transfected cells (Fig. 6h). Whereas we detected no ligands after
immunoprecipitation from controls that had not been transfected DISCUSSION
with receptors, antibodies to the hemagglutinin-tagged forms of We have found that neurotrophic functions in the fruit fly are carried
Toll-6 and Toll-7 copurified DNT2-V5 and DNT1-V5, respectively, out by Toll-7 and Toll-6 binding DNT1 and DNT2, respectively. Toll-6

1254 VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 nature NEUROSCIENCE


a r t ic l e s

and Toll-7 are expressed in the locomotor circuit, including motor In humans, alterations in NF-κB function lead to psychiatric disor-
neurons and interneurons of the embryonic central pattern genera- ders41. Previous reports have shown that Toll-6 and Toll-7 do not
tor and locomotion centers of the adult central brain. By removing activate Drosomycin upon immune challenge, indicating that Toll-6
Toll-6 and Toll-7 function in mutants or adding them in excess, we and Toll-7 do not have innate immunity functions and do not activate
have shown that Toll-6 and Toll-7 are required for normal locomotion NF-κB–Dif in cell types involved in immunity7,8. In future work we
and motor axon targeting, and to maintain neuronal survival. In the plan to elucidate the signaling mechanism downstream of Toll-6 and
absence of Toll-6 and Toll-7 function, at least some of the dying cells Toll-7 in the CNS and, in particular, to determine whether it uses
are HB9+ and Eve+ EL interneurons that normally express the receptors. downstream signal transducers such as MyD88 that are required for
Using genetic interaction analysis, we have shown that Toll-6 and Toll7 the immune and developmental functions of Toll-1. The mammalian
function together with DNT1 and DNT2 in vivo. Using biochemical TLR-8 is required for neurite extension in the neonatal brain, but this
approaches in vitro, in cell culture and in vivo, we have shown that Toll-6 activity is not MyD88 dependent44. Thus, although our data do not
and Toll-7 directly bind DNT2 and DNT1, respectively. Finally, the rela- confirm or refute whether Toll-6 and Toll-7 can signal through the
tive in vivo protein distribution patterns of the ligands and the receptors canonical Toll signaling pathway, they do show that Toll-6 and Toll-7
are consistent with their shared functions. Most importantly, we have function upstream of NF-κB.
shown that Toll receptors underlie neurotrophism in fruit flies, which is This conclusion is supported by several observations reported here.
therefore implemented using a different molecular mechanism from the First, in cell culture, activated forms of Toll-6 and Toll-7 and stimula-
canonical vertebrate mechanism involving p75NTR, Trks and Sortilin. tion with DNT ligands were able to induce NF-κB signaling via Dorsal
Our data show that Toll-6 and Toll-7 have neurotrophic functions in and Dif. Second, in vivo, overexpression of activated Toll-6CY and
the Drosophila CNS matching those of DNT1 and DNT2 (ref. 20). As in Toll-7CY in retinal photoreceptor neurons resulted in the elevation
the mammalian neurotrophin system, these functions are pleiotropic. of Dorsal, Dif and Cactus proteins, as was previously reported for
Mammalian neurotrophin ligands and receptors have functions rang- Toll-145. Third, in vivo, overexpression in neurons of activated Toll-6CY
© 2013 Nature America, Inc. All rights reserved.

ing from maintaining neuronal survival to axon targeting, dendritic and Toll-7CY, like activated Toll10b, rescued the semi-lethality of the
arborization and synaptic transmission, which vary with context, cell spz2 mutation; and conversely, overexpression of activated Toll10b in
type and time16,38,39. For instance, whereas vertebrate neurotrophins neurons rescued the semi-lethality of the DNT1 DNT2 double muta-
and Trk receptors maintain neuronal survival in the peripheral nerv- tion. Our data also show that signaling by Toll-6 and Toll-7 differs in
ous system, they do not have a prominent role in maintaining motor at least some respects from that mediated by Spz–Toll-1. For example,
neuron survival, instead functioning at the neuromuscular junction in cell culture the activation of NF-κB signaling by Toll-6 and Toll-7
in synaptogenesis and synaptic plasticity40. Our data show that Toll-6 was not as strong as that reported by others to be induced by Toll-17,8;
and Toll-7 also have pleiotropic functions, maintaining predominantly and in vivo genetic rescues revealed a specific and stronger relation-
interneuron survival and regulating motor-axon targeting. ship between Toll-6 and Toll-7 and DNT1 and DNT2, compared to
Our data indicate that Toll-7/DNT1 and Toll-6/DNT2 are the most Toll-1. Understanding the molecular mechanisms of Toll-6 and Toll-7
likely ligand-receptor pairs, but there appears to be promiscuity in signaling that underlie the developmental programs that they promote
ligand binding, as at least DNT2 can bind both receptors. This may is a key objective of future research.
also be the case for DNT1, but pure mature DNT1 protein could Notably, NF-κB, p75NTR and Toll receptors are all evolutionar-
not be obtained using the baculovirus system, restricting the tests ily very ancient molecules, present in cnidarians (for example,
that we were able to perform. Such promiscuity may account for the Nematostella); thus, they evolved long before the common ancestor
redundancy between Toll-6 and Toll-7 observed in genetic and func- of flies and humans and since the origin of the nervous and immune
tional tests (for example, compromised locomotion and viability in systems1,46. Of note, the Toll homolog in the worm Caenorhabditis
the double mutants only). It may indicate that in vivo the binding elegans is expressed in neurons and can implement an immune func-
npg

partners might be determined by the relative temporal and spatial tion by means of a behavioral response of pathogen avoidance 47.
distribution patterns of the proteins. Alternatively, it is also conceiv- p75NTR is a member of the tumor necrosis factor receptor superfamily,
able that DNT1 and DNT2 have distinct functions and may bind which is closer to the Tolls than to the Trks48. Toll receptors resemble
each receptor according to functional requirements. DNT1 and DNT2 p75NTR intracellularly, through their ability to activate a downstream
have distinct biochemical properties: whereas DNT2 is consistently signaling pathway resulting in the activation of NF-κB, and Trk recep-
secreted from S2 cells as a mature, cleaved form consisting of the tors in the extracellular ligand-binding module, with a combination
cystine knot domain, DNT1 is secreted both as full-length and mature of leucine-rich repeats and cysteine repeats48. Trk receptors, with an
forms, as well as products of cleavage in the disordered pro-domain. intracellular tyrosine kinase domain, emerged later in evolution 49.
The protease that might cleave DNT1 in vivo is unknown, but these Although Toll receptors are evolutionarily conserved, they are not,
properties are akin for DNT2 to the intracellular cleavage of NGF and at least in the innate immunity context, activated by the same ligands
for DNT1 the extracellular cleavage of BDNF16. In either case, the in flies and humans1. This raises questions: if in Drosophila the Trk
observed promiscuity is reminiscent of the binding of all mammalian receptors were lost and Tolls are the only neurotrophic receptors,
neurotrophins to a common p75NTR receptor. is this a key difference that underlies the distinct brain types and
Although vertebrate neurotrophin receptors are structurally and behaviors in flies and humans? In the course of evolution, did the
functionally distinct from the Tolls, both regulate NF-κB1,13,32,33,41. Tolls become specialized for immunity functions in vertebrates? Or
NF-κB is also one of the transcription factors that activates the innate is the relationship uncovered here between the neurotrophin-ligand
immune response downstream of the TLRs, and it also has extensive and Toll-receptor superfamilies an ancient mechanism of nervous
and highly conserved functions in neurons. Neuronal NF-κB controls system formation? In mammals TLRs also have nervous system func-
gene expression as a potent prosurvival factor; it controls neurite tions, including ones in neurogenesis, neurite growth, plasticity and
extension; it also has non-nuclear synaptic functions, including the behavior, but the endogenous ligands in the mammalian CNS are
clustering of glutamate receptors; and it underlies synaptic plasticity unknown50. A key objective of future research will be to investigate
during learning and memory, from crustaceans to mammals31,32,41–43. whether the neurotrophin and TLR protein families interact in the

nature NEUROSCIENCE VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 1255


a r t ic l e s

mammalian brain, particularly in the context of learning, memory, 15. Gangloff, M. et al. Structural insight into the mechanism of activation of the Toll
and neurodegenerative and neuroinflammatory diseases. receptor by the dimeric ligand Spätzle. J. Biol. Chem. 283, 14629–14635 (2008).
16. Lu, B., Pang, P.T. & Woo, N.H. The yin and yang of neurotrophin action. Nat. Rev.
Neurosci. 6, 603–614 (2005).
Methods 17. Arnot, C.J., Gay, N.J. & Gangloff, M. Molecular mechanism that induces activation
of Spatzle, the ligand for the Drosophila Toll receptor. J. Biol. Chem. 285,
Methods and any associated references are available in the online 19502–19509 (2010).
version of the paper. 18. Hoffmann, A. et al. Biophysical characterization of refolded Drosophila Spatzle,
a cystine knot protein, reveals distinct properties of three isoforms. J. Biol. Chem.
Note: Any Supplementary Information and Source Data files are available in the 283, 32598–32609 (2008).
online version of the paper. 19. Hoffmann, A., Neumann, P., Schierhorn, A. & Stubbs, M.T. Crystallization of Spatzle,
a cystine-knot protein involved in embryonic development and innate immunity in
Acknowledgments Drosophila melanogaster. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 64,
We thank C. Arnot, J. Wen and M. Wheatley for advice; S. Jondhale, J. Ng and 707–710 (2008).
S. Quayle for technical help; S. Bishop and K. Kato for comments on the 20. Zhu, B. et al. Drosophila neurotrophins reveal a common mechanism for nervous
manuscript; A.J. Courey (University of California, Los Angeles), J.L. Imler (Institut system formation. PLoS Biol. 6, e284 (2008).
de Biologie Moléculaire et Cellulaire, CNRS, Strasbourg), T. Ip (University of 21. Parker, J.S., Mizuguchi, K. & Gay, N.J. A family of proteins related to Spatzle, the toll
receptor ligand, are encoded in the Drosophila genome. Proteins 45, 71–80 (2001).
Massachusetts), J.M. Reichhart (Institut de Biologie Moléculaire et Cellulaire,
22. Jacobs, J.R. The midline glia of Drosophila: a molecular genetic model for the
CNRS, Strasbourg), S. Sanyal (Emory University), R. Baines (University of developmental functions of glia. Prog. Neurobiol. 62, 475–508 (2000).
Manchester), M. Freeman (University of Oxford), K. Ito (University of Tokyo), 23. Rogulja-Ortmann, A., Lüer, K., Seibert, J., Rickert, C. & Technau, G.M. Programmed
A. Chiba (University of Miami), B. Pfeiffer (Janelia Farm), M. Landgraf (University cell death in the embryonic central nervous system of Drosophila melanogaster.
of Cambridge), C. Basler (University of Zurich), H. Broihier (Case Western Development 134, 105–116 (2007).
Reserve University), D. Ferrandon (CNRS, Strasbourg), the Bloomington 24. White, K. et al. Genetic control of programmed cell death in Drosophila. Science
Stock Center and Iowa Hybridoma Bank for reagents; the Birmingham Mass 264, 677–683 (1994).
Spectrometry Facility (Birmingham Science City, Advantage West Midlands); 25. Hidalgo, A. et al. Trophic neuron-glia interactions and cell number adjustments in
the fruit fly. Glia 59, 1296–1303 (2011).
and Len Packman for mass spectrometry and Edman sequencing in Cambridge.
26. Palgi, M. et al. Evidence that DmMANF is an invertebrate neurotrophic factor supporting
The LTQ Orbitrap Velos ETD mass spectrometer used in this research was
© 2013 Nature America, Inc. All rights reserved.

dopaminergic neurons. Proc. Natl. Acad. Sci. USA 106, 2429–2434 (2009).
obtained through the Birmingham Science City Translational Medicine: 27. Newquist, G. et al. Blocking apoptosis signaling rescues axon guidance in Netrin
Experimental Medicine Network of Excellence project, with support from mutants. Cell Rep. 3, 595–606 (2013).
Advantage West Midlands (AWM). This work was funded by a UK Medical 28. Bergmann, A., Tugentman, M., Shilo, B.Z. & Steller, H. Regulation of cell number
Research Council Career Establishment Grant (MRCG0200140) to A.H., Wellcome by MAPK-dependent control of apoptosis: a mechanism for trophic survival signaling.
Trust project grant (WT094175/Z/10/Z) to A.H. and N.J.G., Wellcome Trust Dev. Cell 2, 159–170 (2002).
equipment grant (WT073228/Z/03/Z) to A.H., Wellcome Trust programme grant 29. Hidalgo, A., Kinrade, E.F.V. & Georgiou, M. The Drosophila neuregulin vein maintains
glial survival during axon guidance in the CNS. Dev. Cell 1, 679–690 (2001).
(WT081744MA) to N.J.G., European Union Marie Curie International Incoming
30. Learte, A.R., Forero, M.G. & Hidalgo, A. Gliatrophic and gliatropic roles of PVF/PVR
Fellowship (PIIF-GA-2010-274193-NPN) to J.S.W., UK Medical Research Council signaling during axon guidance. Glia 56, 164–176 (2008).
studentship to G.M. and Brunei government studentship to M.A.L. 31. Gutierrez, H. & Davies, A.M. Regulation of neural process growth, elaboration and
structural plasticity by NF-κB. Trends Neurosci. 34, 316–325 (2011).
AUTHOR CONTRIBUTIONS 32. Foehr, E.D. et al. NF-κB signaling promotes both cell survival and neurite
G.M., I.F., J.A., J.S.W., M.A.L., J.C.F. and A.H. performed experiments; A.H. and process formation in nerve growth factor-stimulated PC12 cells. J. Neurosci. 20,
N.J.G. conceived and directed the project; A.H., N.J.G. and G.M. wrote the 7556–7563 (2000).
paper; all authors contributed to planning experiments and analyzing data and 33. Carter, B.D. et al. Selective activation of NF-κB by nerve growth factor through the
to discussions and improvements to the manuscript. neurotrophin receptor p75. Science 272, 542–545 (1996).
34. Levi-Montalcini, R., Aloe, L. & Alleva, E. A role for Nerve Growth Factor in nervous,
COMPETING FINANCIAL INTERESTS endocrine and immune systems. Prog. Neuroendocrinimmunol. 3, 1–10 (1990).
35. Rivest, S. Regulation of innate immune responses in the brain. Nat. Rev. Immunol.
The authors declare no competing financial interests.
9, 429–439 (2009).
36. Sanyal, S. Genomic mapping and expression patterns of C380, OK6 and
Reprints and permissions information is available online at http://www.nature.com/
D42 enhancer trap lines in the larval nervous system of Drosophila. Gene Expr.
reprints/index.html.
Patterns 9, 371–380 (2009).
37. Forero, M.G., Pennack, J.A., Learte, A.R. & Hidalgo, A. DeadEasy caspase: automatic
npg

1. Leulier, F. & Lemaitre, B. Toll-like receptors—taking an evolutionary approach. counting of apoptotic cells in Drosophila. PLoS ONE 4, e5441 (2009).
Nat. Rev. Genet. 9, 165–178 (2008). 38. Huang, E.J. & Reichardt, L.F. Trk receptors: roles in neuronal signal transduction.
2. Janeway, C.A. Jr. & Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. Annu. Rev. Biochem. 72, 609–642 (2003).
20, 197–216 (2002). 39. Blum, R. & Konnerth, A. Neurotrophin-mediated rapid signaling in the central nervous
3. Imler, J.L. & Zheng, L. Biology of Toll receptors: lessons from insects and mammals. system: mechanisms and functions. Physiology (Bethesda) 20, 70–78 (2005).
J. Leukoc. Biol. 75, 18–26 (2004). 40. Liu, X. & Jaenisch, R. Severe peripheral sensory neuron loss and modest motor
4. Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J.M. & Hoffmann, J.A. The neuron reduction in mice with combined deficiency of brain-derived neurotrophic
dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent
factor, neurotrophin 3 and neurotrophin 4/5. Dev. Dyn. 218, 94–101 (2000).
antifungal response in Drosophila adults. Cell 86, 973–983 (1996).
41. Mattson, M.P. & Meffert, M.K. Roles for NF-κB in nerve cell survival, plasticity,
5. Yagi, Y., Nishida, Y. & Ip, Y.T. Functional analysis of Toll-related genes in Drosophila.
and disease. Cell Death Differ. 13, 852–860 (2006).
Dev. Growth Differ. 52, 771–783 (2010).
42. Freudenthal, R. & Romano, A. Participation of Rel/NF-κB transcription factors in
6. Nakamoto, M. et al. Virus recognition by Toll-7 activates antiviral autophagy in
long-term memory in the crab Chasmagnathus. Brain Res. 855, 274–281 (2000).
Drosophila. Immunity 36, 658–667 (2012).
43. Heckscher, E.S., Fetter, R.D., Marek, K.W., Albin, S.D. & Davis, G.W. NF-κB, IκB,
7. Tauszig, S., Jouanguy, E., Hoffmann, J.A. & Imler, J.L. Toll-related receptors
and IRAK control glutamate receptor density at the Drosophila NMJ. Neuron 55,
and the control of antimicrobial peptide expression in Drosophila. Proc. Natl. Acad.
Sci. USA 97, 10520–10525 (2000). 859–873 (2007).
8. Ooi, J.Y., Yagi, Y., Hu, X. & Ip, Y.T. The Drosophila Toll-9 activates a constitutive 44. Ma, Y. et al. Toll-like receptor 8 functions as a negative regulator of neurite outgrowth
antimicrobial defense. EMBO Rep. 3, 82–87 (2002). and inducer of neuronal apoptosis. J. Cell Biol. 175, 209–215 (2006).
9. Seppo, A., Matani, P., Sharrow, M. & Tiemeyer, M. Induction of neuron-specific 45. Reichhart, J.M. et al. Expression and nuclear translocation of the rel/NF-κB-related
glycosylation by Tollo/Toll-8, a Drosophila Toll-like receptor expressed in non-neural morphogen dorsal during the immune response of Drosophila. C.R. Acad. Sci. III
cells. Development 130, 1439–1448 (2003). 316, 1218–1224 (1993).
10. Kambris, Z., Hoffmann, J.A., Imler, J.L. & Capovilla, M. Tissue and stage-specific 46. Putnam, N.H. et al. Sea anemone genome reveals ancestral eumetazoan gene
expression of the Tolls in Drosophila embryos. Gene Expr. Patterns 2, 311–317 (2002). repertoire and genomic organization. Science 317, 86–94 (2007).
11. Hoffmann, J.A. The immune response of Drosophila. Nature 426, 33–38 (2003). 47. Pujol, N. et al. A reverse genetic analysis of components of the Toll signaling
12. Weber, A.N.R. et al. Binding of Drosophila cytokine Spatzle to Toll is direct and pathway in Caenorhabditis elegans. Curr. Biol. 11, 809–821 (2001).
establishes signaling. Nat. Immunol. 4, 794–800 (2003). 48. Bothwell, M. Evolution of the neurotrophin signaling system in invertebrates. Brain
13. Gay, N.J. & Gangloff, M. Structure and function of Toll receptors and their ligands. Behav. Evol. 68, 124–132 (2006).
Annu. Rev. Biochem. 76, 141–165 (2007). 49. Sossin, W.S. Tracing the evolution and function of the Trk superfamily of receptor
14. DeLotto, Y. & DeLotto, R. Proteolytic processing of the Drosophila Spatzle protein tyrosine kinases. Brain Behav. Evol. 68, 145–156 (2006).
by easter generates a dimeric NGF-like molecule with ventralising activity. 50. Okun, E., Griffioen, K.J. & Mattson, M.P. Toll-like receptor signaling in neural
Mech. Dev. 72, 141–148 (1998). plasticity and disease. Trends Neurosci. 34, 269–281 (2011).

1256 VOLUME 16 | NUMBER 9 | SEPTEMBER 2013 nature NEUROSCIENCE


ONLINE METHODS and pAct5C-Pro-TEV6HisV5-DNT2-CK fusion constructs encoding full-length
Genetics. Mutants and reporters. Toll-6MiMICMI02127 encodes a GFP-bearing DNT1 and DNT2. TEV protease sites designed to aid protein cleavage were not
insertion into the coding region of Toll-6 (http://flybase.org/reports/FBti0140037. used, as they cleaved spontaneously. The DNT1 and DNT2 signal peptide and
html). Toll-626, Toll-631, Toll-7P8 and Toll-7P114 are null mutant alleles generated pro-domain sequences were PCR-amplified from DNT1 and DNT2 cDNAs, with
by imprecise excision of P-element insertions (gift of J.L. Imler). Deficiencies primers 27, 28, 33 and 34 (Supplementary Table 3). The DNT1 and DNT2 cys-
Df(3L)DXG4 and Df(2R)BSC22 uncover Toll-6 and Toll-7, respectively. DNT141, tine knots (plus the C-terminal extension CK-CTD for DNT1) were amplified
DNT2e03444 and spz2 have been described20,51. All stocks were balanced using using primers 29, 30, 35 and 36 (Supplementary Table 3). Using overlapping
lacZ-marked balancers and/or TM6B Tb to identify mutants. Drosomycin-GFP PCR, tagged full-length DNT1 and DNT2 sequences were obtained and cloned
(gift of J.M. Reihhart) is a reporter for Dif signaling. using Gateway.
Overexpression in vivo. Overexpression in vivo used the following GAL4 drivers: To produce DNT2 protein using baculovirus-infected Sf9 insect cells, full-
(1) w;;elavGAL4 for all neurons, (2) w;chaGAL4 (gift from R. Baines) for cholinergic length DNT2 was PCR amplified from clone LD26258 (Berkeley Drosophila
neurons, (3) line D42 (gift from S. Sanyal) for Toll-6-GAL4 (ref. 36), (4) w;Toll- Genome Project) using primers 31 and 32 (Supplementary Table 3). The insert
7GAL4, (5) w;GMRGAL4 (gift from M. Freeman) for retina, (6) w;;HB9GAL4 was cloned into the pFastBac1 vector (Invitrogen) using EcoRI and NotI sites.
for HB9-neurons, and (7) w;engrailedGAL4 (Bloomington). These were crossed
to (1) membrane-tethered reporters: (i) w;;UASGAP-GFP (gift from A. Chiba), Protein purification. Purification of Toll-6ECD and Toll-7ECDs produced from
(ii) w;UASmCD8-GFP, (iii) w;;10xUAS-myr-td-Tomato (gift from B. Pfeiffer), S2 cells. S2 cells were transfected with pAct-Toll-6-ECD-6His-3xFLAG for
(iv) UASDsRed (gift from K. Ito), (v) w;Lim3GAL4UASmyrRFP/CyOactYFP Toll-6ECD or pAct-7-ECD-6His-3xFLAG for Toll-7ECD and incubated for
(gift from M. Landgraf), (2) activated forms of the receptors: (i) w;UASToll-6∆LRR 72 h. Protein purification was performed as described above for DNT1 and
and w;UASToll-7∆LRR , (ii) w;;UASToll-6CY and w;;UASToll-7CY, (iii) w;UASToll10b DNT2. Toll-6 and Toll-7 ECDs were identified by mass spectrometry at the
(gift from T. Ip). Other lines were generated by conventional genetics. Proteomics Facility, University of Birmingham. Ni-NTA–purified Toll-6 and
Survival assays. Flies were bred at 18 °C, as stocks or crossed from hetero- Toll-7 ECD proteins were further purified with anti-FLAG magnetic beads
zygous mutants over balancer chromosomes. The survival index (SI) is given by (Sigma-Aldrich) by standard procedures. The purified proteins were used for
SI = 2 × TM6B+/TM6B−. A SI of 1 is the Mendelian expectation when viability native gel electrophoresis.
© 2013 Nature America, Inc. All rights reserved.

is unaffected. See Supplementary Table 2 for genotypes of parental flies and Purification of DNT2 produced from baculovirus expression system. A secreted
sample sizes. DNT2 protein was produced in Sf9 insect cells by baculovirus infection with
the DNT2-Pro-CK-TEV-6xHis sequence. The protein was purified as described
Generation of fusion constructs. Toll-7GAL4. To generate Toll-7GAL4, a 5-kb previously17. Edman (N-terminal) sequencing was carried out at the Protein and
fragment immediately upstream of the Toll-7 start ATG was amplified by PCR Nucleic Acid Chemistry (PNAC) facility at the Department of Biochemistry,
using primers 1 and 2 (Supplementary Table 3) and cloned into 5′ NotI and 3′ University of Cambridge. The cleaved, mature cystine knot domain of DNT2
BamHI restriction sites of the pPTGAL vector. was purified by reverse phase chromatography, as previously described for
Cloning of full-length Toll-6 and Toll-7. Toll-6 and Toll-7 are intronless genes. Spz (ref. 15).
Full-length open reading frames, equivalent to cDNAs, were PCR-amplified from
genomic DNA using primers 3–6 (Supplementary Table 3), and Gateway cloning Mass spectrometry. Toll-6ECD and Toll-7ECD verification. Coomassie bands
was used to generate pAct-Toll-6-HA and pAct-Toll-7-HA fusion constructs. were excised from a gel, destained and subjected to in-gel digestion with trypsin
Cloning of Toll-6ECD-His-FLAG and Toll-7ECD-His-FLAG. Sequences encod- for overnight at 37 °C using standard procedures. Peptides were extracted from
ing the extracellular domains (ECD) of Toll-6 and Toll-7 were cloned to pro- gel pieces with acetonitrile and formic acid and dried in an evaporator. The
duce secreted forms of the ligand-binding domains for binding assays in vitro. samples were resuspended in 0.1% formic acid/water and subjected to liquid
Sequences encoding Toll-6 and Toll-7 ECDs tagged with 6His were PCR- chromatography–tandem mass spectrometry that was performed using an
amplified using primers 7–10 (Supplementary Table 3). Gateway cloning was Ultimate 3000 HPLC series (Dionex) coupled to a LTQ Orbitrap Velos ETD
used to generate pAct-Toll-6-ECD-6His-3xFLAG and pAct-7-ECD-6His- mass spectrometer (ThermoFisher Scientific) via a Triversa Nanomate nanospray
3xFLAG fusion constructs. source (Advion Biosciences). Peptide separation, mass spectrometric analysis and
Generation of activated forms of Toll-6 and Toll-7 for cell culture and database search were carried out as specified at the University of Birmingham
npg

transgenesis. The activated Toll-6 and Toll-7 receptors UASToll-6∆LRR-HA and Proteomics Facility.
UASToll-7∆LRR-HA comprise the signal peptide, transmembrane and intra­cellular DNT2 verification. DNT2CK was analyzed by MALDI-TOF mass spectrom-
domains of Toll-6 and Toll-7, respectively, but lack the entire extracellular etry, following procedures previously described for Spz15.
domain. PCR from cDNA was used to amplify signal peptide sequences (primers
11, 12, 15, 16; Supplementary Table 3) separately from transmembrane- Western blotting. Western blotting was carried out following standard pro­cedures.
intracellular domain sequences (primers 13, 14, 17, 18; Supplementary Table 3), Primary antibodies used were mouse anti-6-His (1:4,000, BD Pharmingen,
and MluI sites were added to each, 3′ and 5′ respectively. MluI sites were used #552565) or mouse anti-6-His (1:1,000, Thermo Scientific, #MA1-21315),
to ligate fragments, which were then cloned into the pUAS-Gateway-HA-attB mouse anti-V5 (1:5,000, Invitrogen, #R960-25), mouse anti-Dorsal (1:500,
destination vector (gift of C. Basler) to produce pUAS-Toll-7∆LRR-HA-attB and Hybridoma Bank, 7A4), mouse anti-Cactus (1:500, Hybridoma Bank, 3H12),
pUAS-Toll-6∆LRR-HA-attB fusion constructs. rabbit anti-Dif (1:500, ref. 52, gift from D. Ferrandon), chicken anti-HA (1:2,000
In UAS-Toll-6CY and UAS-Toll-7CY the conserved cysteines at position 1020 of for S2 cell co-IP western blot; 1:5,000 for fly in vivo co-IP, #ET-HA100) and
Toll-6 and position 993 of Toll-7 are substituted by tyrosine, mimicking the con- mouse anti-HA (12CA5) (1:2,000, Roche, #11 583 816 001). Secondary anti-
stitutively active allele of Toll, Toll10b, and the functional UASToll10b constructs. bodies used were HRP–anti-mouse (1:5,000, Vector Labs, #PI-2000), HRP–anti-
Overlap extension PCR following standard procedures was used to make the rabbit (1:5,000, Vector Labs, #PI-1000) and HRP–anti-chicken (1:10,000, Jackson
cysteine-to-tyrosine mutations in each receptor. The primers used were as follows: ImmunoResearch, #703-035-155). For quantitative analysis of dorsal, cactus and
5′ primers 19, 20, 23 and 24 and 3′ primers 21, 22, 25 and 26 (Supplementary Dif expression, five adult fly heads were pooled per sample (n = 4, total 20 flies
Table 3). The resulting products were cloned into the UAS-Gateway-FLAG des- per genotype), then lysed in NP-40 lysis buffer (50 mM Tris-HCl, pH 8.0,
tination vector to produce pUAS-Toll-7CY-FLAG and pUAS-Toll-6CY-FLAG 150 mM NaCl, 1% NP-40). Western blot images were analyzed by GeneTools
fusion constructs. software (Syngene). Band intensities were normalized to Ponceau red staining
Sequencing confirmed that in UAS-Toll-7CYFLAG and UAS-Toll-6CYFLAG of the same membrane.
the targeted cysteines had been mutated to tyrosines. There is an additional muta-
tion in UAS-Toll-7CYFLAG of Pro58 to leucine and one in UAS-Toll-6CYFLAG S2 cell culture signaling assays. Toll-6 and Toll-7 transfections. DNT1 was
of Ser862 to threonine, both in the extracellular domain. produced in transfected S2 cells (Invitrogen), and DNT2 was produced by both
Cloning for DNT protein production. DNT1 and DNT2 proteins were pro- transfected S2 cells and Sf9 cells infected with baculovirus. Purified ligands were
duced by S2 cell expression, from pAct5C-Pro-TEV6HisV5-DNT1-CK-CTD added to S2 cells transfected with Toll-6 and Toll-7 receptors to test activation of

doi:10.1038/nn.3474 nature NEUROSCIENCE


the snail-luciferase reporter by Dorsal or nuclear translocation of Dorsal; the cell Toll-6ECD and Toll-7ECD were purified from S2 cell conditioned medium,
line 648-1B6 stably transfected with drosomycin-luciferase was used to test the and DNT2-CK was produced using baculovirus infection and purified by reverse
activation of the drosomycin-luciferase reporter by Dif. To test downstream signal- phase chromatography. 12.5 µl Ni-NTA- and FLAG-purified Toll-6 and/or Toll-7
ing by the activated receptors, S2 cells were transfected with 1 µg pMTGAL4 and 1 ECD were mixed with 2 µl of reverse-phase-purified DNT2-CK and left on ice for
µg pUAS-Toll-7CY-FLAG or with 1 µg pUAS-Toll-6CY-FLAG. To test signaling by 30 min to form complexes. Protein mixtures or proteins alone were supplemented
Toll-6 and Toll-7 upon DNT binding, full-length Toll-6 and Toll-7 were expressed with 4× native gel-loading buffer (62.5 mM Tris-HCl, pH 8.8, 20% glycerol and
in S2 cells by transfecting with 1 µg of the pAct-Toll-7-HA or pAct-Toll-6-HA 0.005% bromophenol blue). Samples were separated on a 6% polyacrylamide gel
constructs. S2 cells and the S2 cell line 648-1B6 1B6 stably transfected with for 1 h at 100 V followed by 1 h at 150 V in the absence of SDS. Proteins were then
drosomycin-luciferase were maintained by standard procedures, and the analyzed by western blotting.
TranIT-2020 (Mirus) transfection reagent was used. In all cases, pAct-Renilla
was cotransfected as a control. To determine luciferase activity, we used the Generation of anti–Toll-7, anti-DNT1 and anti-DNT2 antibodies. Antibodies
Dual-Glo Luciferase Assay System (Promega). Cell suspension (50 µl) was trans- were raised to Toll-7 using peptide AAQRAQTWRPKREQLHLQQA injected
ferred in triplicate to an opaque 96-well plate. The supplied firefly luciferase sub- into guinea pigs, and antisera were affinity purified (Davids Biotechnologie).
strate (40 µl) was added, samples were incub­ated for 10 min at room temperature, Antibodies were raised to DNT1 using peptide VRYARPQKAKSASGEWKY
and luminescence was measured using a SPECTRAFluro Plus (Tecan). Stop & and to DNT2 using peptide KRLIALQGNGQN; peptides were injected into
Glo substrate (40 µl) was added, samples were incubated at room temperature rabbits and antisera were affinity purified (Davids Biotechnologie).
for 10 min and luminescence was measured. The relative luciferase activity was
determined by normalizing the firefly over the Renilla readout for each well. In situ hybridizations and immunohistochemistry. In situ hybridizations fol-
lowed standard procedures using mRNA probes, from pDONR-Toll-7 linearized
Coimmunoprecipitation (co-IP). In vivo co-IP from transgenic flies. Transgenic with HindIII and pDONR-Toll-6 linearized with SmaI, both transcribed with
flies of genotype w;GMRGAL4/UASDNT13′+FLAG; UASToll-7HA/+ and wild- T7 RNA polymerase.
type Oregon R control flies were used for in vivo co-IP. Heads were homogenized Immunolabeling was carried out following standard procedures. Primary
in NP40 lysis buffer and spun at 1,200g for 5 min at 4°. Supernatant was precleared antibodies used were rabbit anti-GFP (1:1,000 for embryos and larvae, 1:250
© 2013 Nature America, Inc. All rights reserved.

with 20 µl of protein-A/G magnetic beads (Pierce) in a total volume of 500 µl for for adult brains, Invitrogen #A11122), mouse anti-GFP (1:1,000, Invitrogen
1 h at 4° and then incubated with 1 µg mouse anti-Flag antibody (cat. no. F3165, #A11120), rabbit anti-DsRed (1:100, Clontech #632496), mouse anti-FasII
clone M2, Sigma-Aldrich) overnight at 4 °C. Protein-A/G magnetic beads (25 µl) (1:4 for embryos, 1:250 for larvae, DSHB ID4), mouse anti–tyrosine hydroxylase
were added to the lysate and incubated at room temperature for 1 h. The beads (1:50, Immunostar #22941), rabbit anti–cleaved-Caspase-3 (1:50, Cell Signaling
containing the immune complex were washed twice with lysis buffer and once #9661; 1:250, AbCam #Ab13847), mouse anti–β-gal (1:750, Sigma #G4644),
in PBS. The immune complex was eluted in 2× Laemmli buffer by incubating for guinea pig anti-HB9 (1:1,000, ref. 53 gift from H. Broihier), rabbit anti-HB9
10 min at room temperature and was analyzed by SDS-PAGE. (1:1,000, ref. 53, gift from H. Broihier), mouse anti-Eve (1:5–1:10, DSHB 2B8),
Co-IP from S2 cells. S2 cells were transfected with 1 µg pAct-Toll-6-HA, mouse anti-Dorsal (1:10, DSHB 7A4), guinea pig anti–Toll-7 (see above; 1:10),
pAct-Toll-7-HA, pAct5C-Pro-TEV6HisV5-DNT1-CK-CTD or pAct5C- rabbit anti-DNT1 (see above; 1:50 for adult brains, 1:100 for embryos) and rabbit
Pro-TEV6HisV5-DNT2-CK, or cotransfected with 1 µg pAct5C-Pro- anti-DNT2 (see above; 1:100 for embryos and larvae, 1:50 for adult brains).
TEV6HisV5-DNT1-CK-CTD plus 1 µg pAct-Toll-7-HA per well or with 1 µg Secondary antibodies were directly conjugated Alexa 488, 546 and 647 (1:250,
pAct5C-Pro-TEV6HisV5-DNT2-CK plus 1 µg pAct-Toll-6-HA per well. After Molecular Probes) or biotinylated mouse or rabbit (1:300) followed by avidin
48 h cells were collected and centrifuged at 1,000g for 5 min at 4 °C. After wash- amplification using the Vectastain ABC Elite kit (Vector Labs) or Streptavidin–
ing in PBS, cells were lysed in 600 µl NP-40 lysis buffer with protease inhibitor Alexa 488 (1:250, Molecular Probes). Stainings were carried out in populations
cocktail (Pierce Biotechnology) and left on ice for 30 min. After lysis, samples of hundreds of embryos, at least three adult brains and three larval VNCs per
were centrifuged at 14,000g for 10 min at 4 °C. Five hundred microliters from experiment, and experiments were repeated at least twice and most often more.
each lysate was precleared with 20 µl of protein-A/G magnetic beads (Pierce) for
1 h at 4 °C. Precleared lysates were incubated with 1.5 µg anti-V5 or 1 µg anti- Imaging. Laser-scanning confocal microscopy was carried out using a Leica
HA overnight at 4 °C. Cell lysate–antibody mixtures were incubated with 25 µl SP2-AOBS and a 40× or 63× lens at 512 × 512 or 1,024 × 1,024 pixels resolution,
npg

protein-A/G magnetic beads for 1 h at room temperature. Beads were washed with 0.5- or 1-µm steps. Caspase-positive apoptotic cells in vivo were counted
two to four times in NP-40 lysis buffer and once in PBS. Proteins were eluted in automatically using DeadEasy Caspase software37.
40 µl 2× Laemmli buffer and analyzed by SDS-PAGE.
Automatic tracking of larval locomotion. Larvae were collected from
ELISA. S2 cells were seeded, transfected and processed as described above crosses kept in vials with standard yeast-rich food at 25 °C in a 12 h light:12h dark
for co-IP. ELISA was carried out following standard procedures. High-binding- regimen. All larvae analyzed were also homozygous white (w) mutant. For our
capacity ELISA plate (Greiner Bio-One) wells were coated with anti-V5 (1:1,000) w+;Toll-626 stock, to obtain w;Toll-631/Toll-626 larvae, F1 larvae from a cross to
or anti-HA (1:1,000). Each well was incubated with 100 µl of cell lysate and then w;Toll-631 females were sexed and only w− males were used. Larvae were collected,
with anti-HA (1:1,000) or anti-V5 (1:1,000), respectively. ELISA signal was devel- rinsed in water and placed on a large petri dish containing agar, Vogel-Bonner
oped by TMB substrate (Pierce) and the reaction was stopped with 1 M H3PO4. salts and 40% glucose at room temperature and on a light box. Larvae were
Absorbance was measured at 450 nm. allowed to recover for 10–20 s before filming began and were then filmed for
1 min. All filming was carried out in the morning between 1 and 5 h after zeitgeber
Native gel electrophoresis. To test whether the secreted Toll-6ECD and Toll- ‘lights on’ time, and larvae from all genotypes were filmed in each session. Larvae
7ECD could interact with mature DNT2-CK, proteins were mixed and puta- were filmed using a Motic MC Camera 1.1 or a Canon video camera.
tive complexes were run on a native gel. In native gels, protein mobility does Films were analyzed using FlyTracker software (our modification of the ImageJ
not depend on molecular weight, but on conformation and charge, relative to MTrack2 plug-in). Canon films were first converted to .avi format using Any
the pH of the buffer (as the difference between the buffer pH and the pI of the Video Converter Professional with codec .mjpeg, and Motic films were decom-
proteins increases, the proteins migrate further). We used sample and gel buff- pressed using VirtualDub software (http://www.virtualdub.org/) and saved in .avi
ers at pH = 8.8 and running buffer pH = 8.6. The pI of DNT2CK-TEV-6His is format. Films were checked in VirtualDub for sequence continuity and images
7.94, very close to the buffer pH. The pI of Toll-6ECD-6His-3xFLAG is 6.06 not containing larvae edited out to ensure a continuous sequence of filmed crawl-
and that of Toll-7ECD-6His-3xFLAG is 6.0, quite different from the buffer pH. ing larvae. The first 400 frames were opened blindly in ImageJ and converted
For pI and net charge calculation, we used Protein Calculator v3.3 (http://www. to grayscale, the resolution was set to 800 × 600 pixels, and the frames were
scripps.edu/~cdputnam/protcalc.html). At the pH of our buffers, both proteins saved as a stack of .tiff images. These were next analyzed using FlyTracker in
are negatively charged. ImageJ. FlyTracker tracks the crawling larvae and produces three outputs per film:

nature NEUROSCIENCE doi:10.1038/nn.3474


a stack of 400 images with the location of the larva as identified by the software analyzed with Kruskal-Wallis for the whole data set, followed by Dunn’s test for
in each corresponding raw image; a path or trajectory of each larva in each film multiple comparisons. Categorical data were analyzed with chi-squared tests,
as a single JPEG image; and an Excel spreadsheet with quantitative locomotion followed by a z-test and Bonferroni post hoc to the whole data set or Bonferroni
parameters. The speed in mm/s was converted from the output in pixels using a corrections for pairwise comparisons to controls. No blinding was done. For
photographed reference of known size in mm. All trajectories for each genotype genetic in vivo experiments, the reproducibility of the experiment was verified
were plotted onto a single image using Adobe Photoshop layers. by the overall large population sizes; cell culture experiments were carried out
in general at least three times in triplicate. P values, tests and sample sizes are
Statistical analysis. For continuous data, kurtosis, skewness, histograms and provided in the Results text and figure legends, and further details are provided
Kolmogorov-Smirnov normality tests were carried out to test data distribution, in Supplementary Table 1.
and Levene tests for the homogeneity of variance. For a few samples that were not
distributed normally within a data set that showed normal distribution, normal- 51. Weber, A.N. et al. Role of the Spatzle pro-domain in the generation of an active
ity was assumed. Normally distributed data were analyzed with unpaired t-tests toll receptor ligand. J. Biol. Chem. 282, 13522–13531 (2007).
for two-sample comparisons and one-way ANOVA (or Welch versions for sig- 52. Rutschmann, S. The Rel protein DIF mediates the antifungal but not the antibacterial
host defence in Drosophila. Immunity 12, 569–580 (2000).
nificant Levene tests) for the whole data set followed by Dunnett (fixed control
53. Broihier, H.T. & Skeath, J.B. Drosophila homeodomain protein dHb9 directs neuronal
group) or Games-Howell (significant Levene tests) post hoc tests or Bonferroni fate via crossrepressive and cell-nonautonomous mechanisms. Neuron 35, 39–50
corrections for multiple t-tests. Data that were not distributed normally were (2002).
© 2013 Nature America, Inc. All rights reserved.
npg

doi:10.1038/nn.3474 nature NEUROSCIENCE

You might also like