You are on page 1of 14

J Comp Physiol A (2014) 200:251–264

DOI 10.1007/s00359-014-0891-5

Review

Neuroethology of male courtship in Drosophila: from the gene


to behavior
Daisuke Yamamoto · Kosei Sato ·
Masayuki Koganezawa 

Received: 19 July 2013 / Revised: 29 August 2013 / Accepted: 11 February 2014 / Published online: 25 February 2014
© Springer-Verlag Berlin Heidelberg 2014

Abstract  Neurogenetic analyses in the fruit fly Dros- dTrpA1 Drosophila transient receptor potential
ophila melanogaster revealed that gendered behaviors, cation channel subfamily A1
including courtship, are underpinned by sexually dimor- EcR Ecdysone receptor
phic neural circuitries, whose development is directed in EPSPs Excitatory postsynaptic potentials
a sex-specific manner by transcription factor genes, fruit- fru  fruitless
less (fru) and doublesex (dsx), two core members compos- Gr Gustatory receptor
ing the sex-determination cascade. Via chromatin modifi- HDAC1 Histone deacetylase 1
cation the Fru proteins translated specifically in the male HP1 Heterochromatin protein 1
nervous system lead the fru-expressing neurons to take on IR84a Ionotropic receptor 84a
the male fate, as manifested by their male-specific sur- mAL Medially located just above the antennal
vival or male-specific neurite formations. One such male- lobe
specific neuron group, P1, was shown to be activated Or67d Olfactory receptor 67d
when the male taps the female abdomen. Moreover, when P2X2 ATP-activated P2X purinoceptor 2
artificially activated, P1 neurons are sufficient to induce ppk  pickpocket
the entire repertoire of the male courtship ritual. These SOG Suboesophageal ganglion
studies provide a conceptual framework for understanding TIF1 Transcription intermediary factor 1
how the genetic code for innate behavior can be embodied TNT Tetanus toxin light chain
in the neuronal substrate. tra  transformer

Keywords  fruitless · Sexual dimorphism ·


Identified neurons · Pheromones · Chromatin Introduction

Abbreviations The courtship ritual of Drosophila melanogaster is histori-


7-T 7-Tricosene cally considered as a fixed action pattern that is composed of
7,11-HD 7,11-Heptacosadiene several discrete behavioral elements (Bastock and Manning
bon  bonus 1955; Hall 1982; Greenspan and Ferveur 2000) (Fig. 1a).
cVA  cis-Vaccenyl acetate These sequential actions are indeed genetically programmed
DEG/ENaC Degenerin/epithelial sodium channels and have been dissected by the genetic method. In the past
dsx  doublesex few years, spectacular advances have been made in unrave-
ling the causal links among the genes, circuitry and behaviors
involved in the courtship as fuelled by many newly developed
D. Yamamoto (*) · K. Sato · M. Koganezawa  methodologies such as optogenetic and thermogenetic tools
Division of Neurogenetics, Tohoku University Graduate School
even at single-cell resolution (Luo et al. 2008; Rideout et al.
of Life Sciences, 2‑1‑1 Katahira, Aoba‑ku, Sendai 980‑8577,
Japan 2010; Venken et al. 2011; Ruta et al. 2010; Kohatsu et al.
e-mail: daichan@m.tohoku.ac.jp 2011; Pan et al. 2011; von Philipsborn et al. 2011).

13

252 J Comp Physiol A (2014) 200:251–264

Fig. 1  Courtship behavior and neural sex determination in Drosoph- tion). Copulation usually persists for 15–20 min. At the end of copu-
ila. a Steps (1–6) of courtship behavior and the time of FruM action lation, the pair releases their genital coupling and the male dismounts
in development. When a male finds a moving female, he orients his from the female’s back (disengagement). The inseminated female
body axis toward the female and starts to chase her (orientation and becomes unreceptive for a few days to a week and engages in ovipo-
following). Upon approaching the female, the male fly often touches sition. Fru deficiency in males during the prepupal and pupal stages
her abdomen with his foreleg, on which pheromone-sensitive gus- leads to altered courtship behavior after adult emergence (fru mutant).
tatory organs are distributed (tapping). At the same time, the male b The sex-determination cascade. The sex chromosome composition
fly vibrates, for seconds to minutes, a wing of the side closer to the for female (XX) and male (XY) individuals is translated into the dif-
female while circling around her (wing extension and vibration). ferential transcriptional states, active (ON) and silence (OFF), of the
These unilateral wing vibrations generate so-called courtship songs, primary female-determinant gene Sxl, respectively. The female-spe-
which calm the female (Bennet-Clark and Ewing 1968; von Schil- cific Sxl regulates splicing of its target, tra, ensuring the female-spe-
cher 1976a, b). When a female becomes sufficiently receptive upon cific production of Tra. The presence of Tra represses the production
the exposure to courtship songs, the male licks her genitalia with his of FruM in females, whereas its absence in males allows the produc-
proboscis (licking) and attempts to mount her (attempted copulation). tion of FruM, which masculinizes a population of neurons. Tra also
The receptive female responds to the male by her raising wings and acts on dsx, yielding male-type DsxM and female-type DsxF, each of
opening the vaginal plate, assisting the male in his mounting and, which supports the sex-specific development of neural and non-neural
when his attempt is rewarding, allowing copulation to start (copula- cells

Among dozens of mutant flies isolated, fruitless (fru) noted the complete lack of copulation accompanied by a
mutant flies have opened up a new avenue for the study of strong tendency for homosexual courtship in fru1 males
the molecular and cellular bases of courtship behavior. In (Gill 1963). Phenotypic analyses placed fru downstream of
this review, we discuss the molecular mechanism whereby transformer (tra) (Hall 1977; Gailey and Hall 1989; Gailey
the master regulator Fru instructs the formation of sexually et al. 1991; Taylor 1992; Taylor et al. 1994), a female-deter-
dimorphic neural circuitries and how these circuitries oper- minant gene in the sex determination cascade (Salz 2011)
ate to guide sex-specific courtship behavior. Finally, we (Fig.  1b). Subsequent molecular cloning of the fru gene
attempt to present a general framework which will assist in verified this proposal (Ito et al. 1996; Ryner et al. 1996).
the exploration of the mechanistic basis for sexually dimor- Specifically, the second exon of fru was found to contain
phic behavior in other organisms. three repeats of a 13-nucleotide stretch that matched a Tra-
binding consensus sequence that had been found in the
doublesex (dsx) gene, the only Tra target known at that time
Molecular basis of fruitless function in neural (Nagoshi et al. 1988). Tra is a female-determinant protein
sex determination that is translated only in females and stimulates splicing
near its binding site on the primary RNA transcript (also
Fru sexually dimorphic expression known as precursor mRNA or pre-mRNA) (Nagoshi et al.
1988). Due to the presence or absence of Tra, the fru pri-
The first mutant allele of fru (now called fru1) was iso- mary transcript derived from the most distal promoter is
lated in 1963 by Gill (1963) as a byproduct in screens for subjected to sexually dimorphic splicing, yielding female-
male sterile mutants with impaired spermatogenesis. Gill type transcripts with premature stop codons and male-type

13
J Comp Physiol A (2014) 200:251–264 253

transcripts with long open reading frames (Ryner et al. observation prompted the authors to propose that Fru acts
1996; Heinrichs et al. 1998; Lam et al. 2003). Because of like a master regulator gene in organizing the CNS circuitry
this differential splicing, functional Fru proteins are trans- for male courtship behavior (Demir and Dickson 2005). A
lated in the male but not the female nervous system, where similar result has been reported (Manoli et al. 2005) based
the most distal promoter is active (Usui-Aoki et al. 2000; on the analysis of other fru knock-in alleles.
Lee et al. 2000). The fru gene has at least three additional
promoters that are active in both sexes outside the nervous Sexual dimorphism in single neurons
system (Ito et al. 1996; Ryner et al. 1996). These non-sex-
specific Fru proteins are poorly characterized (Anand et al. A subsequent study (Kimura et al. 2005) documented a con-
2001) and will not be further considered here. In addition to spicuous sexual dimorphism of a cluster of fru-expressing
the second exon, the terminal exon is also subject to alter- interneurons. The cluster was identified using a technique
native splicing, resulting in five different types of C-termini known as Mosaic Analysis with a Repressible Cell Marker
of Fru (Ito et al. 1996; Ryner et al. 1996; Usui-Aoki et al. (MARCM; Lee and Luo 1999), which can be used to label
2000; Goodwin et al. 2000). a group of cells that are derived from a single neuroblast (a
The Fru proteins have a BTB domain near the N-termi- neuroblast clone) or even a single cell (a single-cell clone).
nus and 2 zinc-finger motifs in the C-terminus, with the This cluster, which is “medially located just above the
exception of two isoforms lacking the Zinc-finger portion antennal lobe (mAL)” (Lee et al. 2000), exhibits sex differ-
(Ito et al. 1996; Ryner et al. 1996; Usui-Aoki et al. 2000). ences in three distinct features (Fig. 2a): the cluster com-
Because the BTB domain is involved in protein dimeriza- prises 5 neurons in females and 30 in males; in females,
tion and zinc-fingers are well-known DNA-binding motifs, the neurons have a single contralateral neurite descending
Fru proteins likely contribute to transcriptional regulation to the suboesophageal ganglion (SOG), whereas in males,
that functions only in the male nervous system. The func- some of the neurons have ipsilateral as well as contralat-
tional differences between the five Fru isoforms have not eral neurites; in the SOG, the neurite bifurcates to form
been firmly established (Song et al. 2002; Billeter et al. a Y-shape in females, whereas it terminates in a simple
2006). horsetail-like structure in males. In males with strong loss-
of-function fru mutations, the mAL cluster is feminized
Switching the neural sex by manipulating fru with respect to all three features described above. Wild-
type females have a smaller number of mAL neurons due
Evidence that fru acts as a neural sexual switch was first to active elimination of a proportion of cells, as evidenced
provided by the observation that artificial expression of by the fact that deleting three cell death genes—hid, grim
Fru in a female nervous system leads to the formation of a and reaper—with the deficiency chromosome Df[H99]
male-specific trait in such females. For example, a pair of dramatically increases the number of mAL neurons in the
male-specific muscles (the muscle of Lawrence) (Lawrence female but not the male brain (Kimura et al. 2005; Kimura
and Johnson 1984, 1986; Gailey et al. 1991; Currie and 2011). It is possible that mAL neurons are protected from
Bate 1995; Taylor and Knittel 1995; Nojima et al. 2010) is cell death by Fru in the male brain. The supernumerary
induced when a motoneuron innervating the muscle is mas- mAL neurons that escaped cell death in the female brain by
culinized by the forcible expression of a normal type of fru the action of Df[H99] have the Y-shaped dendritic arbors,
(fru+) from a transgene integrated into an unspecified site retaining the female-typical structure (Kimura et al. 2005),
of the genome (Usui-Aoki et al. 2000). However, expres- but extend an ipsilateral neurite toward the SOG, a male-
sion of fru+ from this transgene in females was unable to specific structure. These observations indicate, first, that
switch the behavioral gender (D. Yamamoto, unpublished the sexual dimorphism in the three features—cell number,
observation), presumably because CNS effects probably the presence or absence of the ipsilateral neurite and the
require a precise balance in the expression of different Fru tip shape of the contralateral neurite—is produced by dis-
isoforms, unlike the simple induction of the male fate in a tinct molecular mechanisms and, second, that Fru controls
muscle by a single motoneuron. all three of these characteristics (Kimura et al. 2005; Goto
One study described how the endogenous fru locus was et al. 2011).
engineered with the aim to generate an entire set of Fru- Exhaustive identification of other fru-expressing neurons
encoding transcripts that can be expressed regardless of by subsequent studies using the MARCM or intersectional
the sex of the animal (Demir and Dickson 2005). Females technique revealed structural sex differences in approxi-
homozygous for this constitutive fru allele (fruM) displayed mately one-third of ~2,000 fru-expressing neurons (Cash-
male-typical courtship behavior toward other females, ero et al. 2010; Yu et al. 2010; Chiang et al. 2011). The
although this courtship activity was not as strong as that development of methods to scale individual neurons on a
seen in wild-type males (Demir and Dickson 2005). This standardized brain (Jefferis et al. 2007; Chiang et al. 2011)

13

254 J Comp Physiol A (2014) 200:251–264

Fig. 2  Mechanism of action of
Fru in the production of sex dif-
ferences in single neurons of the
mAL cluster. a The mAL cluster
exhibits sex-related differences
in cell number, the presence or
absence of ipsilateral neurites,
and the shape of contralateral
neurites. b The mAL cluster in
males carrying a hypomorphic
fru mutation has an intersexual
appearance. Single-cell clones
that carry the fru hypomorphic
mutation showed either a male-
typical or female-typical struc-
ture (left-hand side the result
was consistent with hypothesis
1) and did not show an inter-
sexual structure (right-hand side
the result was consistent with
hypothesis 2), indicating an all-
or-none mode of the sex deter-
mination at the single-cell level.
c Single mAL neurons adopt
either a female fate or a male
fate depending on the amount
of Fru they express. d Changes
in chromatin factor expression
phenocopy fru mutants in the
mAL neuron development. e
Proposed mechanism by which
Fru and interacting factors alter
the expression of target genes
that promote or inhibit neural
masculinization

has enabled researchers to deduce the presence of connec- phenotype is regulated by genetic modifiers, as shown in
tions among neighboring arbors of different neurons, and experiments in which heterozygous loss-of-function alleles
this has resulted in a putative circuit diagram in which fru- of 435 different genes with a known function in neural
expressing neurons are found to connect with each other development were introduced. These studies showed that in
(Yu et al. 2010), although it needs to be verified whether the case of 14 genes, a loss-of-function allele enhanced fru-
these putative connections are actually functional. induced roughness, and in the case of 4 genes, a loss-of-
function alleles of 4 genes suppressed it (Goto et al. 2011;
Molecular mechanism of Fru action Ito et al. 2012). One of the enhancers thus obtained was an
Ecdysone receptor mutation (Goto et al. 2011), which had
To obtain insights into the molecular function of Fru, previously been reported to synergistically interact with fru
genetic screens were conducted for mutations that modify mutant alleles to enhance male–male courtship and abro-
the action of Fru (Goto et al. 2011). The results showed gate neural sexual dimorphism in the antennal lobe (Dalton
that overexpression of a normal type of fru (fru+) in the et al. 2009). A potent suppressor of the fru-induced rough-
developing eye disc distorts the regular alignment of the eye phenotype was a loss-of-function allele in bonus (bon)
ommatidia (unit eyes in the compound eye), leading to (Ito et al. 2012). Bon is a fly homolog of the mammalian
the so-called rough-eye phenotype. The severity of this transcription intermediary factor 1 family of transcriptional

13
J Comp Physiol A (2014) 200:251–264 255

cofactors, which are known to participate in gene silenc- is tempting to hypothesize that, in masculinizing a neuron,
ing by associating with histone deacetylase 1 (HDAC1) Fru recruits HDAC1 to the target sites for inducing chro-
and heterochromatin protein 1 (HP1) (Nielsen et al. 1999; matin condensation and gene silencing and that this process
Schreiber and Bernstein 2002). The involvement of Bon is fine-tuned by HP1a, which counteracts it (Fig. 2e).
and these chromatin factors in fru-dependent development
of the sex-specific circuitry was suggested by genetic inter-
actions between the respective mutations; the reduced male Cellular basis of Fruitless function in courtship circuit
courtship in hypomorphic fru mutants was dominantly development
enhanced by a loss-of-function allele in bon or HDAC1 (on
the genetic locus Rpd3) and alleviated by a loss-of-function The male‑specific P1 neuron cluster
allele in HP1a (the genetic locus Su(var)205) (Ito et al.
2012). Thus, all these factors likely interact in fru-depend- Sex differences are prevalent among fru-expressing neu-
ent sexual development of the nervous system. However, rons. Some are sexually dimorphic as is the mAL cluster
rather unexpectedly, HDAC1 and HP1a appear to counter- described above. Others are sex-specific, including the
act each other, i.e., HDAC1 normally supports Fru-depend- male-specific P1 cluster (Fig. 3a, b). This cluster consists
ent masculinisation of the nervous system, whereas HP1a of 20 interneurons that are located in the dorsal posterior
supports de-masculinization (Ito et al. 2012) (see also Liu brain and that have a trans-midline neurite connecting lat-
et al. 2005; Shirangi et al. 2006). eral protocerebra (Fig. 3c). The P1 cluster plays a central
The hypothesis stemming from these behavioral obser- role in initiating male courtship. In a behavioral applica-
vations was supported by the examination of single-neu- tion of the MARCM technique (Kimura et al. 2008), tra−
ron anatomy (Fig. 2b). In males carrying hypomorphic mutant clones were generated in the female brain to exam-
fru mutations, the mAL cluster (visualized en masse as a ine whether tra−-induced masculinization of a particular
neuroblast clone) exhibits intersexual characteristics: it subset of fru-expressing neurons is sufficient for a female
has ~10 cells, ipsilateral neurites and Y-shaped branches to exhibit male-like courtship behavior. Out of ~200 tra−
in the contralateral terminal. However, single-cell clones mosaic females, 16 females courted a female, using a male-
of mAL cluster neurons show no intersexual characteris- like behavioral pattern. In 13 of these 16 females (81 %)
tics, i.e., each mAL neuron is either male-type or female- the P1 neuron cluster had been masculinized unilaterally or
type. Interestingly, in such fru hypomorphic males, knock bilaterally (Fig. 3d3). For another ~40 fru-expressing clus-
down of HDAC1 increases the proportion of male-type ters assessed in this study, there was no correlation between
cells, whereas knock down of HP1a decreases it. Therefore, masculinization and the occurrence of male-like behavior
HDAC1 drives masculinization of single neurons and HP1a in the tra− mosaic female flies. Thus, the P1 cluster is a
impedes it. This indicates that, in these neurons, Fru func- potent activator of male-type courtship and may function
tions as an all-or-none sexual switch (Fig. 2c), in which as a decision-making center that initiates this behavior
HDAC1 and HP1a have opposing actions to each other (Ito (Kimura et al. 2008).
et al. 2012) (Fig. 2e). The P1 cluster is not present in females unless the femi-
Immunostaining of polytene chromosomes has revealed nizing factor tra is inactivated, as in the MARCM flies used
that Fru binds approximately 130 locations. Bon binds to 90 in the experiment described above (Kimura et al. 2008). As
Fru-target sites and an additional ~100 locations. HDAC1 in the case of the mAL cluster (Kimura et al. 2005), the P1
binds to 86 out of 90 Fru-Bon labeled sites and HP1a to 20 cluster is absent in females as a result of the female-specific
of these sites, including several sites shared by HDAC1 and death of neurons (Kimura et al. 2008). However, unlike in
HP1a. The binding of HDAC1 and HP1a to the Fru target the case of mAL, Fru proteins have no role in regulating
sites is abrogated in the absence of Bon (Fru itself remains the male-specific presence of the P1 cluster. Instead of fru,
to bind to these sites), indicating that Bon is required for another Tra target, dsx, is the key factor here. The female-
binding of HDAC1 and HP1a to the Fru target sites (Ito specific product of this gene, DsxF, eliminates the P1 clus-
et al. 2012). Immunoprecipitation assays demonstrated that ter from the female brain (Kimura et al. 2008; Kimura
Bon, HDAC1 and HP1a are contained in the Fru-protein 2011). In fact, approximately 65 neurons (Rideout et al.
complex and HDAC1 and HP1a compete with each other 2007, 2010) (out of ~150 dsx-positive neurons) (Lee et al.
in the recruitment to this complex (Ito et al. 2012). As 2002; Rideout et al. 2007, 2010; Sanders and Arbeitman
HDAC1 mediates histone deacetylation that silences tran- 2008; Robinett et al. 2010) in the brain express both fru and
scription (Schreiber and Bernstein 2002; Kharchenko et al. dsx and the P1 cluster is composed of this type of neuron
2010; The modENCODE Consortium 2010) and HP1 has (Kimura et al. 2008). Consistent with this, females carry-
been shown to play a role in both silencing and activation ing a knock-in fru allele, fruM, lack the P1 cluster because
of euchromatic gene transcription (Piacentini et al. 2003), it they have the DsxF protein, yet these females display

13

256 J Comp Physiol A (2014) 200:251–264

a d1

male

d2

female

d3

c female carrying P1 neurons


artificially induced

d4

male whose P1 neurons are


activated by TrpA1

Fig. 3  P1 neurons with the ability to initiate male courtship. Neu- by an arrow. d The correlation of the P1 neurons with the male court-
ronal staining with a GAL4 enhancer trap that drives expression in ship initiation. d1 Wild-type males carrying a pair of P1 cluster in the
the P1 cluster highlights the presence and absence of the P1 cluster brain courted a female. d2 Wild-type females lacking the P1 cluster
in the male a and female b brain, respectively. The somata of P1 neu- in the brain did not court a female. d3 Females in which a P1 cluster
rons in the male brain are circled in (a) and the corresponding region was artificially produced by MARCM courted a female in a pattern
of the female brain is circled in (b). c A neuroblast clone of the P1 similar to that of males. d4 Solitary males that carried the dTrpA1
cluster labeled by MARCM. The somata of P1 neurons are circled. transgene in the P1 cluster initiated courtship when dTrpA1 was acti-
The midline of the brain is indicated with a broken line and the con- vated by a temperature increase
tralateral arbors associated with the trans-midline neurites are shown

male-type courtship behavior toward females (Kimura warmth-activated channel, in a subset of neurons—using
et al. 2008). This means that, even though the P1 cluster the MARCM technique or the intersectional GAL4-UAS
strongly promotes the initiation of male-type courtship, strategy (Luo et al. 2008)—induces firing in these neu-
it is not absolutely necessary for this behavior to occur. rons when the ambient temperature was increased (Venken
Another implication of results of the behavioral MARCM et al. 2011). This approach has revealed two sets of neurons
approach is that the female neural network is able to pro- that can induce courtship behavior in a male placed alone,
duce motor outputs for male-type courtship when driven to i.e., without any courtship target (Kohatsu et al. 2011; Pan
do so by, for example, ectopically producing the P1 cluster et al. 2011; von Philipsborn et al. 2011). One of the two
in the female brain as has been shown in females carrying sets of neurons was the P1 cluster (Fig. 3d4). The other set
tra− neurons. of neurons, which includes P2b (Kohatsu et al. 2011) and
pIP10 (von Philipsborn et al. 2011) (Fig. 4a), comprised
P1 neurons initiate male courtship fru-expressing descending interneurons with terminals in
the thoracic ganglia, where the presumptive motor pattern
Forced activation or inactivation of a subset of neurons is a generator for courtship exists. pIP10 is a single neuron,
powerful technique to determine their functions in behav- whereas P2b consists of ~10 neurons (Kohatsu et al. 2011;
ior. Transgenic expression of transient receptor poten- von Philipsborn et al. 2011). It is tempting to postulate
tial cation channel subfamily A member 1 (dTrpA1), a that the P2b–pIP10 set functions as a command center that

13
J Comp Physiol A (2014) 200:251–264 257

Fig. 4  The core neural com-


a
ponents that drive a male to
sing. a A diagram showing the
presumptive circuitry for male
courtship. b The male court-
P1: Decision making center
ship song. c A tethered male
fly placed on a treadmill (upper
panel) was subjected to neural
activity recordings with a Ca2+
indicator expressed in fru-
positive neurons (lower panel).
P2b: Command fiber
d The fly shown in c was
stimulated by a female contact,
which resulted in a Ca2+ rise
in the lateral protocerebrum in
the brain dPR1: Song generator c

vPR6: IPI regulator

vMS11: Output gate

d
Sine song Pulse song

drives the thoracic motor pattern generator when activated induced in P1 neurons when the tethered male contacted,
by the P1 cluster neurons, whose arborizations are closely by a foreleg, a glass rod coated with an extract of body
apposed to P2b and pIP10 dendrites in the lateral protocer- surface hydrocarbons, which are major contact-chemical
ebrum (Fig. 4a). pheromones, whereas touching an uncoated glass rod did
Physiological correlates of the courtship-initiating capa- not induce any response (Kohatsu et al. 2011). Indeed,
bility of P1 neurons were recorded from a behaving fly by receptors for contact chemicals (gustatory receptors) are
means of optical imaging with a Ca2+ sensor protein, Yel- distributed in leg sensory hairs, some of which contain
low cameleon, expressed specifically in all fru-expressing pheromone-responsive cells (see below). These observa-
neurons (Fig. 4c, d) or P1 MARCM clones (Kohatsu et al. tions support the notion that the activation of the P1 neu-
2011). For this purpose, the authors invented the ‘teth- rons by chemosensory cues initiates courtship behavior in
ered male’ preparation in which a male fly is anchored to males.
a metal wire on his dorsal thorax and is forced to grasp a
Styrofoam ball on which he “walks” while remaining in
a stationary position. This allows neural activity record- Ethological significance of input–output neural
ings of the brain through a window opened on the head regulation
capsule. The tethered male will readily start to chase a
moving female presented in front of him, provided that he Motor control of courtship song
has touched the female with his foreleg (tapping). While
chasing the female, the tethered male typically extends and The decision to court made by P1 neurons is conveyed,
vibrates a wing (Kohatsu et al. 2011), a behavior regarded presumably by P2b/pIP10 neurons, to the song pattern gen-
as a hallmark of male courtship. Touching the female (but erator, which is demonstrated to be located in the thoracic
not seeing a moving female) induced a large Ca2+ transient ganglia (Clyne and Miesenböck 2008). Clyne and Miesen-
in the P1 neurons in these males. Ca2+ transients were böck (2008) showed that decapitated male flies generate

13

258 J Comp Physiol A (2014) 200:251–264

quasi-normal courtship songs composed of pulse and sine pattern generator. The vMS11 neurons are located in the
songs when fru-expressing neurons are forcibly excited via mesothoracic ganglion and innervate the ipsilateral wing
ATP-activated P2X purinoceptor 2 channels that have been neuropile. vMS11 activation via dTrpA1 results in wing
transgenically introduced. These flies show a strong ten- extension without vibration and its inactivation via TNT
dency to use the same wing in successive courtship bouts, abolishes or disturbs song generation. Unilateral wing
in contrast to wild-type males, which typically use two extension is correlated with ipsilateral vMS11 activation.
wings alternately (Clyne and Miesenböck 2008). In addi- Based on these observations, it has been proposed (von
tion, compared to songs of normal male flies (Fig. 4b), the Philipsborn et al. 2011) that vMS11 determines the side on
songs generated by decapitated flies have longer intervals which patterned motor outputs are released into a periph-
between pulses in pulse songs and lower frequency oscil- eral nerve to move a wing and that dPR1 and vPR6 could
lations in sine songs (Pan et al. 2011). The inter-pulse directly receive a descending command from the brain and
interval is unaffected by thorax-restricted fru knockdown be involved in patterning of motor outputs by, for example,
in intact (non-decapitated) males (Rubinstein et al. 2010). determining the inter-pulse interval of pulse songs (Fig. 4).
Female flies (which do not express functional Fru proteins)
also generate courtship song-like sounds, but they are dis- Gustatory inputs in male courtship
torted in pattern (Clyne and Miesenböck 2008; Kohatsu
et al. 2011; Pan et al. 2011; von Philipsborn et al. 2011). The courtship motor pattern generator is controlled by the
Together, these findings indicate, first, that the motor pat- central decision center, in particular, P1 neurons. P1 neu-
tern generator for male courtship songs resides in the tho- rons are activated by pheromone stimulation given to the
racic ganglia of both males and females; and second, that male’s foreleg. What are the pheromones and how are
the brain is dispensable for the motor pattern generation per they perceived by the flies? The composition of cuticular
se, but signals descending from the brain normally modu- hydrocarbons in D. melanogaster is sexually dimorphic;
late the thoracic pattern generator. The signals descending 7-tricosene (7-T) is much more abundant in males than in
from the brain also seem to be necessary for alternate use females and acts as a male-derived aphrodisiac for females
of the two wings and boosting up of the song pitch. Note (and an anti-aphrodisiac for males), whereas 7,11-heptaco-
that although the male-specific P1 cluster is a strong driver sadiene (7,11-HD) is predominantly produced in females
of male courtship initiation, the thoracic motor center can and acts as a female-derived aphrodisiac for males (Jal-
generate song motor outputs if it is activated by some other lon 1984; Billeter et al. 2009; Yew et al. 2009; Everaerts
means regardless of the presence or absence of P1 and P1 et al. 2010). Both compounds are non-volatile and per-
activity. ceived by gustatory receptors in the legs and the mouth
Then which neurons constitute the song pattern genera- part. Exposure to 7-T at physiological concentrations of
tor? The mesothoracic ganglion contains a sexually dimor- 10−12–10−8  M evokes impulses in L2 gustatory neurons in
phic cluster of interneurons expressing both fru and dsx the mouth part of male flies (Lacaille et al. 2007), and stim-
(Rideout et al. 2007, 2010; von Philipsborn et al. 2011). ulation with 7,11-HD induced impulses in the “Tm4c” sen-
Some of these neurons extend arbors in the “wing neuro- sillum on the leg of a male fly (Toda et al. 2012). Further-
pile”, which contains dendrites of motoneurons innervat- more, a study using G-CaMP-aided Ca2+ imaging revealed
ing wing muscles. dTrpA1-induced stimulation of a subset that one of two cells paired in a single sensory hair on the
of these neurons known as vPR6 (Fig. 4a) drives the male leg of a male fly responds to 7-T and the other responds
to sing, and stronger activation with higher temperature to 7,11-HD (Thistle et al. 2012). These pheromone-respon-
increases the pulse rate (i.e., it shortens the inter-pulse sive sensory cells are defined by the presence of pickpocket
interval). Conversely, blocking synaptic transmission in (ppk)-23 and, possibly, ppk-29 expression (Toda et al.
vPR6 through targeted expression of tetanus toxin light 2012; Thistle et al. 2012). ppk genes encode a family of
chain (TNT) prevented the male from singing. The other ion channels known as Degenerin/epithelial sodium chan-
fru and dsx double-positive interneuron cluster is located nels and are expressed in receptor cells for various sensory
in the prothoracic ganglion (Rideout et al. 2007, 2010; modalities. Mutations in ppk23, ppk25 and ppk29 and/
von Philipsborn et al. 2011). One of these neurons, named or knock down of any of these genes reduce male court-
dPR1 (Fig. 4a), produces pulse songs when activated with ship activities toward a female (Lin et al. 2005; Toda et al.
dTrpA1 and inhibits male singing when inactivated through 2012; Thistle et al. 2012; Liu et al. 2012; Lu et al. 2012;
TNT expression. Activation of dPR1 had no effect on the Starostina et al. 2012). Remarkably, sensory neurons that
inter-pulse interval. dPR1 is likely male-specific and it express ppk23, ppk25 and/or ppk29 in the leg sensilla
bilaterally innervates the wing neuropile in the mesotho- also express fru (Lin et al. 2005; Lacaille et al. 2007; Yew
racic ganglion. Another fru-expressing neuron, vMS11 et al. 2009; Toda et al. 2012; Thistle et al. 2012; Liu et al.
(Fig.  4a), has been identified as a component of the song 2012; Lu et al. 2012; Starostina et al. 2012). In females,

13
J Comp Physiol A (2014) 200:251–264 259

the axons of these fru-positive gustatory neurons terminate Gustatory pheromone signal integration
exclusively in the ipsilateral thoracic neuromere, whereas
in males the axons cross the midline and also form termi- Blocking synaptic outputs of Gr32a-expressing gustatory
nals on the contralateral side (Mellert et al. 2010; Thistle neurons disrupts the male courtship posture (Koganezawa
et al. 2012; Liu et al. 2012; Lu et al. 2012; Starostina et al. et al. 2010). Specifically, males with inactivated Gr32a-
2012). The functional importance of this midline crossing expressing neurons are not able to retain a wing in a resting
in males is unknown, despite the fact that this conspicu- position while they are vibrating the other wing, resulting in
ous sexual dimorphism was reported nearly 2 decades ago bilateral wing extension (Koganezawa et al. 2010), in con-
(Possidente and Murphey 1989). It is also unknown how trast to the unilateral wing extension observed in courting
these pheromone inputs from the leg sensilla are relayed to wild-type males. This anomaly is most evident under dark
the higher centers to initiate male courtship. (red light) conditions (Koganezawa et al. 2010), probably
because visual cues can normally compensate for the lack
Gustatory receptors implicated in pheromone perception of pheromone cues in these male flies. The Grs on the left
and right forelegs may sense an asymmetry in the concen-
It remains to be determined whether Ppk family proteins in tration of pheromones on the ground spilled by a female.
the leg sensory cells function as receptors that bind phero- Notably, in males, the axon terminals of Gr32a sensory
mone ligands. Instead, some of the members of the gusta- neurons are juxtaposed to the mAL contralateral neurites in
tory receptor (Gr) family, which is distinctly different from the SOG, whereas in females these two neuronal endings
the Ppk family, have been suggested to function as recep- are spatially separated. This finding leads to the postula-
tors for contact-chemical pheromones in Drosophila. Grs tion that the sex difference in mAL dendritic arborization
are seven-pass transmembrane proteins with no homology patterns is correlated with sexually distinct ways to inte-
to vertebrate chemosensory receptors. Grs are expressed grate pheromone inputs (Koganezawa et al. 2010). mAL
in gustatory neurons locating primarily in the mouth part, interneurons are GABAergic and mutual inhibitory connec-
leg and wing margin (Bray and Amrein 2003; Lacaille tions between the left and right pairs are expected because
et al. 2007; Ejima and Griffith 2008; Moon et al. 2009; arbors of the lateral pairs superimpose and mutual synaptic
Koganezawa et al. 2010; Watanabe et al. 2011). Although connections are not rare in insects. Such lateral inhibition
Gr68a was initially reported to be a candidate receptor would enhance the left–right asymmetry in ascending out-
for female-derived pheromones (Bray and Amrein 2003), puts of mALs, which terminate in a so-called “lateral trian-
subsequent studies supported the alternative possibility gle” in the lateral protocerebrum and a tract in the superior
that Gr68a predominantly functions in mechanosensory medial protocerebrum, the regions where P1 dendrites are
neurons (Ejima and Griffith 2008). Gr39a, a close rela- densely distributed (Kimura et al. 2008). It remains to be
tive of Gr68a, however, might be a receptor for a female- seen whether mAL outputs affect the excitability of P1 or
derived pheromone, as its inactivation curtails male court- P2b neurons.
ship toward a female (Watanabe et al. 2011). Several Gr
proteins have been shown to play roles in inhibiting male Olfactory pathways in male courtship
courtship. Gr33a is likely a co-receptor that presumably
forms complexes with several Grs to constitute functional Although gustatory pheromones play a primary role in
receptors in bitter-sensing cells (Moon et al. 2009). Gr33a Drosophila courtship, olfactory pheromones are also
mutant males engage in male–male courtship at a higher important for successful mating. Major olfactory organs
rate than wild-type males do. In addition, after remov- in Drosophila are the antenna and maxillary palp. The
ing Gr66a-expressing bitter cells, 7-T loses its inhibitory olfactory receptor neurons project to the primary olfac-
effect on male courtship, resulting in an increased male– tory center antennal lobe, which is composed of approxi-
male courtship (Lacaille et al. 2007). Similarly, knockout mately 50 glomeruli, similar to their counterparts in the
of the bitter-taste receptor Gr32a leads to an elevated level mammalian olfactory bulb (Jefferis et al. 2007). The pro-
of male–male courtship (Miyamoto and Amrein 2008). jection neurons extend their axons toward the lateral horn,
These observations are compatible with the idea that some with or without making en passant synapses on the mush-
of the bitter-taste receptors are activated by binding of room body, a multisensory integration center (Jefferis et al.
contact-chemical pheromones. Although these receptors 2007). Three dorsolateral glomeruli, DA1, VA1v and VL2a,
were shown to prevent males from courting other males in are enlarged in males compared to females (Kondoh et al.
D. melanogaster, they might also function to inhibit male 2003; Stockinger et al. 2005). In male moths (e.g., Man-
courtship toward females of other species that have dif- duca sexta) and cockroaches (e.g., Periplaneta americana),
ferent pheromone compositions (Billeter et al. 2009; Fen a few dorsolateral glomeruli form a macroglomerular com-
et al. 2013). plex that represents a hotline for integrating sex pheromone

13

260 J Comp Physiol A (2014) 200:251–264

information (Hildebrand 1996). DA1, VA1v and VL2a glo- generated by females to locate them for courtship, and inhi-
meruli in Drosophila may form a similar pheromone hot- bition of Gr68a-expressing mechanosensory cells impairs
line, segregated from the general odor pathway (Kondoh this ability of males (Ejima and Griffith 2008). By contrast,
et al. 2003). Intriguingly, fru-positive olfactory receptor under light conditions female tracking by a male is guided
neurons selectively innervate these three sexually dimor- primarily by vision (Sakai et al. 1997), although males do
phic glomeruli (Stockinger et al. 2005). not court moving females unless they are primed by phe-
cis-Vaccenyl acetate (cVA) is a volatile pheromone that romonal stimulation (Kohatsu et al. 2011). However, if the
is synthesized in the male accessory gland and transferred courtship ‘decision maker’ P1 is artificially activated by
to the female during copulation, resulting in attenuation of dTrpA1, the male initiates courtship of a moving female, or
her sexual attractiveness, i.e., acting as an anti-aphrodisiac even a moving rubber band, in the absence of pheromonal
for males (Butterworth. 1969; Everaerts et al. 2010). The stimulation (Pan et al. 2012) (only courtship directed to
sexually dimorphic, fru-positive glomerulus DA1 receives females is considered here). It is notable that a population
innervation of olfactory receptor neurons expressing olfac- of fru-positive visual interneurons in the lobular plate inter-
tory receptor 67d, which responds to cVA (Kurtovic et al. digitates with DC1 dendrites (Ruta et al. 2010), implying
2007). The projection neurons from the DA1 glomerulus that they make synaptic connections. Input–output relations
innervate a sexually dimorphic terminal region in the lat- of fru-positive interneurons in the male-specific superior
eral horn (Datta et al. 2008), where they form functional medial protocerebrum tract and lateral triangle are thus key
connections. Indeed, DA1 stimulation induces postsynaptic to understanding the multimodal integration underpinning
excitatory responses in at least three groups of fru-express- courtship decisions.
ing third-order interneurons, DC1, LC1 and LC2, which are Figure  5 shows the model which integrates the experi-
male-specific or sexually dimorphic (Kohl et al. 2013; Ruta mental data on the circuitry, pheromones and receptors
et al. 2010). DC1 interneurons evoke excitatory postsynap- involved in male courtship.
tic potentials (EPSPs) in a fourth-order descending neuron,
DN1, which is male-specific (Ruta et al. 2010). The DN1
neuron extends an axon to the thoracic and abdominal gan- Conclusions and perspectives
glia, implying that it plays a direct role in controlling motor
outputs (Ruta et al. 2010). If this is indeed the case, it is Studies on a mating behavior mutant, fru, in a model organ-
surprising that the inhibitory pheromone cVA can modu- ism, D. melanogaster, successfully unveiled the logic by
late motor output at such a late step of integration (i.e., by which a dedicated single gene orchestrates the genome-
provoking discrete EPSPs in the fourth-order descending wide transcriptional states and ultimately organizes the
neuron). sexually dimorphic neuronal circuitry underlying gendered
behavior. Several recent papers have reported the success-
Multimodal regulation of courtship behavior ful perturbation of mating behavior by the inhibition of
fru with the aid of RNA interference in cockroaches and
Male courtship is not only regulated by pheromones, but grasshoppers, suggesting that fru also instructs the forma-
also by other sensory stimuli, including food odors, sounds tion of sex-specific circuitries for gendered behavior in
and visual stimuli. The antennal lobe glomerulus VL2a these insects. It is an open question whether the same logic
is innervated by olfactory receptor neurons that express underlies the translation of the genetic code for sex-specific
Ionotropic Receptor 84a (IR84a), which responds to food- behavior into the operation principle of the neural circuitry
derived odorous compounds such as phenylacetic acid and in a wide variety of animals. In fact, the existence of a fru
phenylacetaldehyde (Grosjean et al. 2011). In general, the ortholog in mammals remains controversial. However,
neural pathway for processing pheromones is segregated Sry, the male determinant in many mammals (Sekido and
from that for general odors including food odors, each pro- Lovell-Badge 2009), is expressed in the brain in addition
jecting to a different region in the lateral horn. Although to the gonad (Bocklandt and Vilain 2007), the established
VL2a responds to the food odors, the VL2a PNs project to site of its masculinizing action. As Sry is suggested to act
the pheromone integration region in the lateral horn (Gros- through chromatin modification, this finding invites the
jean et al. 2011). Intriguingly, these food odors accelerate speculation that this gene plays a role analogous to fru in
male courtship in an IR84a-dependent manner (Grosjean the mammalian brain.
et al. 2011). Courtship behavior is highly diversified among species,
Male courtship also relies on audition and vision (Eberl presumably because it is exposed to strong pressure of sex-
et al. 1997; Sakai et al. 1997; Ewing 1978; Kamikouchi ual selection. How are these species differences in courtship
et al. 2009; Krstic et al. 2009; Kim et al. 2012; Tootoo- behavior established? Comparative studies with Drosophila
nian et al. 2012). Under dark conditions, males use noise species on neural substrates for courtship behavior will make

13
J Comp Physiol A (2014) 200:251–264 261

DA1 DC1
DA1
Or67d
VA1v DC2
cVA Or47b
LH
LC1
VL2a
IR84a LC2

AL

Gr68a
AMMC
Antenna
mAL P1

7-T Gr32a
Gr33a Gr39a?
Gr66a SOG P2b
Song
PG
DN1
7,11-HD ppk23
ppk25 ppk29 LPR

Foreleg TG Brain

Fig. 5  The input and output channels that regulate male courtship. antennal and mechanosensory motor center, LH lateral horn, SOG
Indicated at the left of the figure are pheromones known to regulate suboesophageal ganglion, TG thoracic ganglia, LPR lateral protocer-
male courtship. Suggested receptors for pheromones are shown in ebrum, Song PG song pattern generator. The spheroids drawn with
the boxes at left, which represent the sensory organs that house the broken lines indicate glomeruli in the antennal lobe. Red boxes indi-
pheromone receptors. The boxes in the middle and at right represent cate fru-positive neurons, whereas black boxes represent fru-negative
neural centers, in which neuron groups involved in the regulation of neurons. Lines represent neurites, whose input and output sites are
courtship behavior are indicated by abbreviations. The squares drawn shown with circles and arrowheads, respectively
with broken lines indicate neuropiles, i.e., AL antennal lobe, AMMC

it possible to determine which differences in the identified the Drosophila melanogaster sex determination gene fruitless.
neurons are critically responsible for species differences in a Genetics 158:1569–1595
Bastock M, Manning A (1955) The courtship of Drosophila mela-
given behavior and which genetic changes in a given gene in nogaster. Behaviour 8:85–111
evolution are causally associated with the behavioral differ- Bennet-Clark HC, Ewing AW (1968) The wing mechanism involved
ences. Rapid advances in the techniques for genetic manipu- in the courtship of Drosophila. J Exp Biol 49:117–128
lation in nonmodel organisms, such as those using TALEN Billeter JC, Villella A, Allendorfer JB, Dornan AJ, Richardson M,
Gailey DA, Goodwin SF (2006) Isoform-specific control of
or CRISPR (Gaj et al. 2013), will open an avenue for the male neuronal differentiation and behavior in Drosophila by the
evo-devo-neuro approach to the question as to how behaviors fruitless gene. Curr Biol 16:1063–1076
were diversified in these species at the molecular and cellular Billeter JC, Atallah J, Krupp JJ, Millar JG, Levine JD (2009) Special-
levels (Yamamoto and Ishikawa 2013). ized cells tag sexual and species identity in Drosophila mela-
nogaster. Nature 461:987–991
Bocklandt S, Vilain E (2007) Sex differences in brain and behavior:
Acknowledgments  The authors’ work is funded by Grants-in- hormones versus genes. Adv Genet 59:245–266
Aid for Scientific Research (24113502, 23220007, 1802012 to D.Y., Bray S, Amrein H (2003) A putative Drosophila pheromone receptor
25132702 and 24700309 to K.S. and 24570082 and 23115702 to expressed in male-specific taste neurons is required for efficient
M.K.) from MEXT, the Strategic Japanese-French Cooperative Pro- courtship. Neuron 39:1019–1029
gram from JST (D.Y.) and a Life Science Grant from the Takeda Sci- Butterworth FM (1969) Lipids of Drosophila: a newly detected lipid
ence Foundation (D.Y. and K.S.). The authors thank M. Suyama and in the male. Science 163:1356–1357
S. Abe for secretarial assistance. Cashero S, Ostrovsky AD, Yu JY, Dickson BJ, Jefferis GS (2010) Sex-
ual dimorphism in the fly brain. Curr Biol 20:1589–1601
Chiang AS, Lin CY, Chuang CC, Chang HM, Hsieh CH, Yeh CW,
References Shih CT, Wu JJ, Wang GT, Chen YC, Wu CC, Chen GY, Ching
YT, Lee PC, Lin CY, Lin HH, Wu CC, Hsu HW, Huang YA,
Anand A, Villella A, Ryner LC, Carlo T, Goodwin SF, Song HJ, Gai- Chen JY, Chiang HJ, Lu CF, Ni RF, Yeh CY, Hwang JK (2011)
ley DA, Morales A, Hall C, Baker BS, Taylor BJ (2001) Molec- Three-dimensional reconstruction of brain-wide wiring net-
ular genetic dissection of the sex-specific and vital functions of works in Drosophila at single-cell resolution. Curr Biol 21:1–11

13

262 J Comp Physiol A (2014) 200:251–264

Clyne JD, Miesenböck G (2008) Sex-specific control and tuning of Ito H, Fujitani K, Usui K, Shimizu-Nishikawa K, Tanaka S, Yama-
the pattern generator for courtship song in Drosophila. Cell moto D (1996) Sexual orientation in Drosophila is altered by
133:354–363 the satori mutation in the sex-determination gene fruitless that
Currie DA, Bate M (1995) Innervation is essential for the develop- encodes a zinc finger protein with a BTB domain. Proc Natl
ment and differentiation of a sex-specific adult muscle in Dros- Acad Sci USA 93:9687–9692
ophila melanogaster. Development 121:2549–2557 Ito H, Sato K, Koganezawa M, Ote M, Matsumoto K, Hama C, Yama-
Dalton JE, Lebo MS, Sanders LE, Sun F, Arbeitman MN (2009) moto D (2012) Fruitless recruits two antagonistic chromatin
Ecdysone receptor acts in fruitless-expressing neurons to medi- factors to establish single-neuron sexual dimorphism. Cell
ate Drosophila courtship behaviors. Curr Biol 19:1447–1452 149:1327–1338
Datta SR, Vasconcelos ML, Ruta V, Luo S, Wong A, Demir E, Flo- Jallon JM (1984) A few chemical words exchanged by Drosophila
res J, Balonze K, Dickson BJ, Axel R (2008) The Drosophila during courtship and mating. Behav Genet 14:441–478
pheromone cVA activates a sexually dimorphic neural circuit. Jefferis GS, Potter CJ, Chan AM, Marin EC, Rohlfing T, Maurer CR
Nature 452:473–477 Jr, Luo L (2007) Comprehensive maps of Drosophila higher
Demir E, Dickson BJ (2005) fruitless splicing specifies male court- olfactory centers: spatially segregated fruit and pheromone rep-
ship behavior in Drosophila. Cell 121:785–794 resentation. Cell 128:1187–1203
Eberl DF, Duyk GM, Perrimon NA (1997) Genetic screen for muta- Kamikouchi A, Inagaki HK, Effertz T, Hendrich O, Fiala A, Gopfert
tions that disrupt an auditory response in Drosophila mela- MC, Ito K (2009) The neural basis of Drosophila gravity-sens-
nogaster. Proc Natl Acad Sci USA 94:14837–14842 ing and hearing. Nature 458:165–171
Ejima A, Griffith LC (2008) Courtship initiation is stimulated by Kharchenko PV, Alekseyenko AA, Schwartz YB, Minoda A, Rid-
acoustic signals in Drosophila melanogaster. PLoS ONE dle NC, Ernst J, Sabo PJ, Larschan E, Gorchakov AA, Gu T,
3:e3246 Linder-Basso D, Plachetka A, Shanower G, Tolstorukov MY,
Everaerts C, Farine J, Cobb M, Ferveur J (2010) Drosophila cuticular Luquette LJ, Xi R, Jung YL, Park RW, Bishop EP, Canfield
hydrocarbons revisited: mating status alters cuticular profiles. TK, Sandstrom R, Thurman RE, MacAlpine DM, Stamatoy-
PLoS ONE 5:e9607 annopoulos JA, Kellis M, Elgin SC, Kuroda MI, Pirrotta V,
Ewing A (1978) The antenna of Drosophila as a ‘love song’ receptor. Karpen GH, Park PJ (2010) Comprehensive analysis of the
Physiol Entomol 3:33–36 chromatin landscape in Drosophila melanogaster. Nature
Fen P, Manoli DS, Ahmed OM, Chen Y, Agarwal N, Kwong S, Cai 471(7339):480–485
AG, Neitz J, Rsnsio A, Baker BS, Shah NM (2013) Genetic and Kim WJ, Jan LY, Jan YN (2012) Contribution of visual and circadian
neural mechanisms that inhibit Drosophila from mating with neural circuits to memory for prolonged mating induced by
other species. Cell 154:89–102 rivals. Nat Neurosci 15:876–883
Gailey DA, Hall JC (1989) Behavior and cytogenetics of fruitless in Kimura K (2011) Role of cell death in the formation of sexual dimor-
Drosophila melanogaster: different courtship defects caused by phism in the Drosophila central nervous system. Develop
separate, closely linked lesions. Genetics 121:773–785 Growth Differ 53:236–244
Gailey DA, Taylor BJ, Hall JC (1991) Elements of the fruitless locus Kimura K, Ote M, Tazawa T, Yamamoto D (2005) Fruitless specifies
regulate development of the muscle of Lawrence, a male- sexually dimorphic neural circuitry in the Drosophila brain.
specific structure in the abdomen of Drosophila melanogaster Nature 438:229–233
adults. Development 113:879–890 Kimura K, Hachiya T, Koganezawa M, Tazawa T, Yamamoto D
Gaj T, Gersbach CA, Barbas CF 3rd (2013) ZFN, TALEN, and (2008) Fruitless and Doublesex coordinate to generate male-
CRISPR/Cas-based methods for genome engineering. Trends specific neurons that can initiate courtship. Neuron 59:759–769
Biotechnol 31:397–405 Koganezawa M, Haba D, Matsuo T, Yamamoto D (2010) The shap-
Gill KS (1963) A mutation causing abnormal courtship and mating ing of male courtship posture by lateralized gustatory inputs to
behavior in males of Drosophila melanogaster. Am Zool 3:507 male-specific interneurons. Curr Biol 20:1–8
Goodwin SF, Taylor BJ, Villella A, Foss M, Ryner LC, Baker BS, Kohatsu S, Koganezawa M, Yamamoto D (2011) Female contact acti-
Hall JC (2000) Aberrant splicing and altered spatial expres- vates male-specific interneurons that trigger stereotypic court-
sion patterns in fruitless mutants of Drosophila melanogaster. ship behavior in Drosophila. Neuron 69:498–508
Genetics 154:725–745 Kohl J, Ostrovsky AD, Frechter S, Jefferis GSXE (2013) A bidirec-
Goto J, Mikawa Y, Koganezawa M, Ito H, Yamamoto D (2011) Sexu- tional circuit switch reroutes pheromone signals in male and
ally dimorphic shaping of interneuron dendrites involves the female brains. Cell 155:1610–1623
Hunchback transcription factor. J Neurosci 31:5454–5459 Kondoh Y, Kaneshiro KY, Kimura K, Yamamoto D (2003) Evolution
Greenspan RJ, Ferveur J (2000) Courtship in Drosophila. Annu Rev of sexual dimorphism in the olfactory brain of Hawaiian Dros-
Genet 34:205–232 ophila. Proc R Soc Lond B 270:1005–1013
Grosjean Y, Rytz R, Farine JP, Abuin L, Cortot J, Jefferis GS, Benton Krstic D, Boll W, Noll M (2009) Sensory integration regulating male
R (2011) An olfactory receptor for food-derived odors promotes courtship behavior in Drosophila. PLoS ONE 4:e4457
male courtship in Drosophila. Nature 478:236–240 Kurtovic A, Widmer A, Dickson BJ (2007) A single class of olfac-
Hall JC (1977) Portions of the central nervous system controlling tory neurons mediates behavioral responses to Drosophila sex
reproductive behavior in Drosophila melanogaster. Behav pheromone. Nature 446:542–546
Genet 7:291–312 Lacaille F, Hiroi M, Twele R, Inoshita T, Umemoto D, Maniere G,
Hall JC (1982) Genetics of the nervous system in Drosophila. Q Rev Marion-Poll F, Ozaki M, Francke W, Cobb M, Everaerts C, Tan-
Biophys 15:223–479 imura T, Ferveur JF (2007) An inhibitory sex pheromone tastes
Heinrichs V, Ryner LC, Baker BS (1998) Regulation of sex-specific bitter for Drosophila males. PLoS ONE 8:e661
selection of fruitless 5′ splice sites by transformer and trans- Lam BJ, Bakshi A, Ekinci FY, Webb J, Graveley BR, Hertel KJ
former-2. Mol Cell Biol 18:450–458 (2003) Enhancer-dependent 5′-splice site control of fruitless
Hildebrand JG (1996) Olfactory control of behavior in moths: central pre-mRNA splicing. J Biol Chem 278:22740–22747
processing of odor information and the functional significance Lawrence PA, Johnson P (1984) The genetic specification of pattern
of olfactory glomeruli. J Comp Physiol A 178:5–19 in a Drosophila muscle. Cell 36:775–782

13
J Comp Physiol A (2014) 200:251–264 263

Lawrence PA, Johnson P (1986) The muscle pattern of a segment of Rideout EJ, Dornan AJ, Neville MC, Eadie S, Goodwin SF (2010)
Drosophila may be determined by neurons and not by contribu- Control of sexual differentiation and behavior by the doublesex
tion myoblasts. Cell 45:505–513 gene in Drosophila melanogaster. Nat Neurosci 13:458–466
Lee T, Luo L (1999) Mosaic analysis with a repressible neurotech- Robinett CC, Vaughan AG, Knapp J, Baker BS (2010) Sex and the
nique cell marker for studies of gene function in neuronal mor- single cell. II. There is a time and place for sex. PLoS ONE
phogenesis. Neuron 22:45–461 8:e1000365
Lee G, Foss M, Goodwin SF, Carlo T, Taylor BJ, Hall JC (2000) Spa- Rubinstein CD, Rivlin PK, Hoy RR (2010) Genetic feminization of
tial, temporal, and sexually dimorphic expression patterns of the the thoracic nervous system disrupts courtship song in male
fruitless gene in the Drosophila central nervous system. J Neu- Drosophila melanogaster. J Neurogenet 24:234–245
robiol 15:404–426 Ruta V, Datta SR, Vasconcelos ML, Freeland J, Looger LL, Axel R
Lee G, Hall JC, Park JH (2002) doublesex gene expression in the cen- (2010) A dimorphic pheromone circuit in Drosophila from sen-
tral nervous system of Drosophila melanogaster. J Neurogenet sory input descending output. Nature 468:686–690
16:229–248 Ryner LC, Goodwin SF, Castrillon DH, Anand A, Villella A, Baker
Lin H, Mann KJ, Starostina E, Kinser RD, Pikilny CW (2005) A BS, Hall JC, Taylor BJ, Wasserman SA (1996) Control of male
Drosophila DEG/ENaC channel subunit is required for male sexual behavior and sexual orientation in Drosophila by the
response to female pheromones. Proc Natl Acad Sci USA fruitless gene. Cell 87:1079–1089
102:12831–12836 Sakai T, Isono K, Tomaru M, Oguma Y (1997) Light-affected male
Liu L, Ni JQ, Shi YD, Oakeley EJ, Sun FL (2005) Sex-specific role following behavior is involved in light-dependent mating in
of Drosophila melanogaster HP1 regulating chromatin structure Drosophila melanogaster. Genes Genet Syst 72:275–281
and gene transcription. Nat Genet 37:1361–1366 Salz HK (2011) Sex determination in insects: a binary decision based
Liu T, Starostina E, Vijayan V, Pikielny CW (2012) Two Drosophila on alternative splicing. Curr Opin Genet Dev 21:395–400
DEG/ENaC channel subunits have distinct functions in gusta- Sanders LE, Arbeitman MN (2008) Doublesex establishes sexual
tory that active male courtship. J Neurosci 32:11879–11889 dimorphism in the Drosophila central nervous system in an
Lu B, LaMora A, Sun Y, Welsh MJ, Ben-Shahar Y (2012) ppk23- isoform-dependent manner by directing cell number. Dev Biol
Dependent chemosensory functions contribute to courtship 132:448–457
behavior in Drosophila melanogaster. PLoS Genet 8:e1002587 Schreiber SL, Bernstein BE (2002) Signaling network model of chro-
Luo L, Callaway EM, Svoboda K (2008) Genetic dissection of neural matin. Cell 111:771–778
circuits. Neuron 57:634–660 Sekido R, Lovell-Badge R (2009) Sex determination and SRY: down
Manoli DS, Foss M, Villella A, Taylor BJ, Hall JC, Baker BS (2005) to a wink and a nudge? Trends Genet 25:19–29
Male-specific fruitless specifies the neural substrates of Dros- Shirangi TR, Taylor BJ, McKeown M (2006) A double-switch system
ophila courtship behavior. Nature 436:395–400 regulates male courtship behavior in male and female Drosoph-
Mellert DJ, Knapp J, Manoli DS, Meissner GW, Baker BS (2010) ila melanogaster. Nat Genet 38:1435–1439
Midline crossing by gustatory receptor neuron axons is regu- Song HJ, Billeter JC, Reynaud E, Carlo T, Spana EP, Perrimon N,
lated by Fruitless, Doublesex and Roundabout receptors. Devel- Goodwin SF, Baker BS, Taylor BJ (2002) The fruitless gene
opment 137:323–332 is required for the proper formation of axonal tracts in the
Miyamoto T, Amrein H (2008) Suppression of male courtship by a embryonic central nervous system of Drosophila. Genetics
Drosophila pheromone receptor. Nat Neurosci 11:874–876 162:1703–1724
Moon SJ, Lee Y, Jiao Y, Montell CA (2009) Drosophila gustatory Starostina E, Liu T, Vijayan V, Zheng Z, Siwicki KK, Pikielny CW
receptor essential for aversive taste and inhibiting male-to-male (2012) A Drosophila DEG/ENaC subunit functions specifically
courtship. Curr Biol 19:1623–1627 in gustatory neurons required for male courtship behavior. J
Nagoshi RN, McKeown M, Burtis KC, Belote JM, Baker BS (1988) Neurosci 32:4665–4674
The control of alternative splicing at genes regulating sexual Stockinger P, Kvitsiani D, Rotkopf S, Tirián L, Dickson BJ (2005)
differentiation in D. melanogaster. Cell 53:229–236 Neural circuitry that governs Drosophila male courtship behav-
Nielsen AL, Ortiz JA, You J, Oulad-Abdelghani M, Khechumian R, Gan- ior. Cell 121:795–807
smuller A, Chambon P, Losson R (1999) Interaction with mem- Taylor BJ (1992) Differentiation of a male-specific muscle in Dros-
bers of the heterochromatin protein 1 (HP1) family and histone ophila melanogaster does not require the sex-determining genes
deacetylation are differentially involved in transcriptional silenc- doublesex or intersex. Genetics 132:179–191
ing by members of the TLF1 family. EMBO J 18:6385–6395 Taylor BJ, Knittel LM (1995) Sex-specific differentiation of a male-
Nojima T, Kimura K, Koganezawa M, Yamamoto D (2010) Neuronal specific abdominal muscle, the Muscle of Lawrence, is abnor-
synaptic outputs determine the sexual fate of postsynaptic tar- mal in hydroxyurea-treated and in fruitless male flies. Develop-
gets. Curr Biol 20:836–840 ment 121:3079–3088
Pan Y, Robinett CC, Baker BS (2011) Turning males on: activation Taylor BJ, Villella A, Ryner LC, Baker BS, Hall JC (1994) Behavioral
of male courtship behavior in Drosophila melanogaster. PLoS and neurobiological implications of sex-determining factors in
ONE 6:e21144 Drosophila. Dev Genet 15:275–296
Pan Y, Messner GW, Baker BS (2012) Joint control of Drosophila The modENCODE Consortium (2010) Identification of functional
male courtship behavior by motion cues and activation of male- elements and regulatory circuits by Drosophila modENCODE.
specific P1 neurons. Proc Natl Acad Sci USA 109:10065–10070 Science 330:1787–1796
Piacentini L, Fanti L, Berloco M, Perrini B, Pimpinelli S (2003) Het- Thistle R, Cameron P, Ghorayshi A, Dennison L, Scott K (2012) Con-
erochromatin protein (HP1) is associated with induced gene tact chemoreceptors mediate male–male repulsion male–female
expression in Drosophila euchromatin. J Cell Biol 161:707–714 attraction during Drosophila courtship. Cell 149:1140–1151
Possidente DR, Murphey RK (1989) Genetic control of sexually Toda H, Zhao X, Dickson BJ (2012) The Drosophila female aphrodis-
dimorphic axon morphology in Drosophila sensory neurons. iac pheromone actives ppk23 + sensory neurons to elicit male
Dev Biol 132:448–457 courtship behavior. Cell Rep 1:599–607
Rideout EJ, Billeter J, Goodwin SF (2007) The sex-determination Tootoonian S, Coen P, Kawai R, Murthy M (2012) Neural represen-
genes fruitless and doublesex specify a neural substrate required tations of courtship song in the Drosophila brain. J Neurosci
for courtship song. Curr Biol 17:1473–1478 32:787–798

13

264 J Comp Physiol A (2014) 200:251–264

Usui-Aoki K, Ito H, Ui-Tei K, Takahashi K, Lukacsovich T, Awano Watanabe K, Toba G, Koganezawa M, Yamamoto D (2011) Gr39a, a
W, Nakata H, Piao ZF, Nilsson EE, Tomida J, Yamamoto D highly diversified gustatory receptor in Drosophila, has a role in
(2000) Formation of the male-specific muscle in female Dros- sexual behavior. Behav Genet 41:746–753
ophila by ectopic fruitless expression. Nat Cell Biol 2:500–506 Yamamoto D, Ishikawa Y (2013) Genetic and neural bases for spe-
Venken KJT, Simpson JH, Bellen HJ (2011) Genetic manipulation of cies-specific behavior in Drosophila species. J Neurogenet. doi:
genes and cells in the nervous system of the fruit fly. Neuron 10.3109/01677063.2013.800060
72:202–230 Yew JY, Dreisewerd K, Luftmann H, Muthing J, Pohlentz G, Krav-
von Philipsborn AC, Liu T, Yu JY, Masser C, Bidaye SS, Dickson BJ itz EA (2009) A new male sex pheromone and cuticular
(2011) Neuronal control of Drosophila courtship song. Neuron cues for chemical communication in Drosophila. Curr Biol
69:509–522 19:1245–1254
von Schilcher F (1976a) The function of pulse song and sine song Yu JY, Kanai MI, Demir E, Jefferis GS, Dickson BJ (2010) Cellular
in the courtship of Drosophila melanogaster. Anim Behav organization of the neural circuit that drives Drosophila court-
24:622–625 ship behavior. Curr Biol 20:1602–1614
von Schilcher F (1976b) The role of auditory stimuli in the courtship
of Drosophila melanogaster. Anim Behav 24:18–26

13

You might also like