Neurons display a striking degree of functional and morphological diversity, and the developmental mechanisms that underlie diversification are of significant interest for understanding neural circuit assembly and function. We find that the morphology of Drosophila sensory neurons is diversified through a series of suppressive transcriptional interactions involving the POU domain transcription factors Pdm1 (Nubbin) and Pdm2, the homeodomain transcription factor Cut, and the transcriptional regulators Scalloped and Vestigial. Pdm1 and Pdm2 are expressed in a subset of proprioceptive sensory neurons and function to inhibit dendrite growth and branching. A subset of touch receptors show a capacity to express Pdm1/2, but Cut represses this expression and promotes more complex dendritic arbors. Levels of Cut expression are diversified in distinct sensory neurons by selective expression of Scalloped and Vestigial. Different levels of Cut impact dendritic complexity and, consistent with this, we show that Scalloped and Vestigial suppress terminal dendritic branching. This transcriptional hierarchy therefore acts to suppress alternative morphologies to diversify three distinct types of somatosensory neurons.

The morphologies of neurons are highly diverse, fitting with distinct functions and connectivity. Transcriptional regulators drive neuronal diversification, as demonstrated by studies in several systems, including rodents, worms and flies (Allan and Thor, 2015). Given the enormous cellular diversity in the nervous system, understanding how multiple transcriptional regulators interact to drive subclass and individual neuronal features is an important goal. Studies of the specification of motor neurons in fly and vertebrate systems have revealed multiple strategies for generating specific axonal trajectories including combinatorial codes, hierarchical genetic cascades, and spatial and temporal patterning (Landgraf and Thor, 2006; Dasen and Jessell, 2009; Enriquez et al., 2015). Drosophila multidendritic sensory neurons have provided a powerful model for identifying transcriptional programs that underlie the morphological diversification of dendritic arbors; nevertheless, regulators have been studied largely in isolation and the strategies for diversification are poorly understood (Corty et al., 2009; Jan and Jan, 2010). Here, we examine transcriptional strategies for the diversification of multiple morphologically and functionally distinct types of somatosensory neurons.

Drosophila sensory neurons are segregated into morphological classes distinguished by sensory dendrite branching pattern, end organ innervation, and axonal targeting in the central nervous system (Bodmer and Jan, 1987; Merritt and Whitington, 1995; Grueber et al., 2002, 2007). A subset of md neurons, the dendritic arborization (da) neurons, extend sensory dendrites across the body wall to cover large territories. The da neurons have been further divided into classes I-IV based on dendritic morphology (Grueber et al., 2002) (Fig. 1A). Dendritic morphology correlates with sensory function, with class I, dbd and dmd1 neurons functioning as proprioceptors (Hughes and Thomas, 2007), class III neurons mediating responses to gentle touch (Yan et al., 2013), and class IV neurons functioning as multimodal nociceptors (Hwang et al., 2007; Xiang et al., 2010). Class II neurons are touch sensitive, but a specific functional role has not been resolved (Tsubouchi et al., 2012).

Multiple transcription factors are known to play a role in sensory neuron dendrite morphogenesis (Moore et al., 2002; Grueber et al., 2003; Li et al., 2004; Sugimura et al., 2004; Kim et al., 2006; Hattori et al., 2007; Jinushi-Nakao et al., 2007; Crozatier and Vincent, 2008; Ye et al., 2011; Ferreira et al., 2014). The transcriptional regulator Cut is a key factor in the establishment of diverse da neuron morphologies (Grueber et al., 2003; Jinushi-Nakao et al., 2007; Iyer et al., 2013). Cut promotes the development of class II, III and IV da neurons with larger territories and complex branching patterns. By contrast, class I, dbd and dmd1 neurons with smaller fields and less complex morphologies lack detectable Cut expression and do not require Cut for dendritic morphogenesis. Cut is expressed at different levels in different da neuron classes. These levels help to specify the development of distinct neuronal morphologies (Grueber et al., 2003). Cut expression is promoted by the Longitudinals lacking (Lola) transcription factor in class IV neurons (Ferreira et al., 2014), but it is still not known how distinct Cut levels are established in different cell types and, more generally, how class-specific expression patterns of transcriptional regulators are generated to diversify sensory neuron morphology.

We addressed these questions by examining links between Cut and the transcription factors Pdm1/2, Scalloped and Vestigial in sensory neuron subset diversification. Our results identify a transcriptional pathway for diversification of sensory neuron morphology via suppression of distinct alternative morphological states.

Cut is required for proper dendritic elaboration and axonal targeting in a subset of da sensory neurons

The homeodomain transcription factor Cut is expressed in class II (low expression), III (high expression) and IV (intermediate expression) da neurons. Prior studies of Cut in da neuron dendritic morphogenesis identified two very distinct phenotypes in cut null mutant MARCM clones. Depending on the specific neuron examined, cut clones either showed simplified dendritic terminal branching or a more severe dendritic growth defect (Grueber et al., 2003). We examined the basis for these qualitatively distinct phenotypes. Neurons that showed severe growth defects included the class II and class III neurons ddaA, ddaB, ddaF and ldaB in the dorsal and lateral clusters (Fig. 1B-F, Fig.2G-K, Fig. S1A,B). Mutant neurons either extended a single primary dendrite that ended in a compact, dense arborization a short distance from the cell body, or adopted a bipolar dendrite morphology (Fig. 1B-D). Stunted dendrites often targeted to a nearby chordotonal organ or nerve (Fig. 1F,F′, Fig. S1B′).

Fig. 1.

Cut function in class II and III dorsal cluster sensory neuron dendrite and axon morphogenesis. (A) Representative tracings and descriptions of peripheral sensory neuron subtypes found in the dorsal cluster of third instar Drosophila larvae. (B) Wild-type class III neuron ddaA MARCM clone. Arrowheads indicate axons here and in subsequent panels. (C) Example of a cutc145 mutant ddaA clone showing stunted dendritic arbor (arrow). (D) Example of a cutc145 mutant ddaA clone displaying a bipolar dendritic morphology (arrows). (E) Wild-type class III ddaF clone. (F,F′) cutc145 mutant ddaF clone (green) showing targeting of stunted dendrites (white arrows) to the v'ch1 chordotonal organ (labeled with HRP, magenta; yellow arrows). (G) Wild-type class III ddaA axons project to the ventral medial (VM) fascicle of the VNC. (Top) Confocal projection of VNC showing ddaA axon (arrow) terminating near the medial fascicles (Fas2, magenta). (Bottom) Transverse view shows axon termination near the VM fascicle (arrow). Fascicle labels: L, lateral; I, intermediate; M, medial. (H) Axon projection of cutc145 mutant ddaA neuron. Top: Axon (arrow) projects to the medial fascicles. Bottom: Transverse view shows aberrant termination near the dorsal medial (DM) fascicle (arrow). (I) Classification of cut mutant axon phenotypes. The percentage of ddaA, ddaB, ddaF and ldaB clones that projected to either dorsal or ventral neuropil regions is shown (classes displayed as group; n=20). (J) Schematic summary of cut axon phenotypes. Wild-type (WT) ddaA, ddaF, ldaB and ddaB axons (green) normally terminated near the VM fascicles. When mutant for cut the axons of these cells (red) instead terminated near the DM fascicles. Fas2+ tracts are shown in magenta. Scale bars: 50 µm in B-F′; 10 µm in G,H.

Fig. 1.

Cut function in class II and III dorsal cluster sensory neuron dendrite and axon morphogenesis. (A) Representative tracings and descriptions of peripheral sensory neuron subtypes found in the dorsal cluster of third instar Drosophila larvae. (B) Wild-type class III neuron ddaA MARCM clone. Arrowheads indicate axons here and in subsequent panels. (C) Example of a cutc145 mutant ddaA clone showing stunted dendritic arbor (arrow). (D) Example of a cutc145 mutant ddaA clone displaying a bipolar dendritic morphology (arrows). (E) Wild-type class III ddaF clone. (F,F′) cutc145 mutant ddaF clone (green) showing targeting of stunted dendrites (white arrows) to the v'ch1 chordotonal organ (labeled with HRP, magenta; yellow arrows). (G) Wild-type class III ddaA axons project to the ventral medial (VM) fascicle of the VNC. (Top) Confocal projection of VNC showing ddaA axon (arrow) terminating near the medial fascicles (Fas2, magenta). (Bottom) Transverse view shows axon termination near the VM fascicle (arrow). Fascicle labels: L, lateral; I, intermediate; M, medial. (H) Axon projection of cutc145 mutant ddaA neuron. Top: Axon (arrow) projects to the medial fascicles. Bottom: Transverse view shows aberrant termination near the dorsal medial (DM) fascicle (arrow). (I) Classification of cut mutant axon phenotypes. The percentage of ddaA, ddaB, ddaF and ldaB clones that projected to either dorsal or ventral neuropil regions is shown (classes displayed as group; n=20). (J) Schematic summary of cut axon phenotypes. Wild-type (WT) ddaA, ddaF, ldaB and ddaB axons (green) normally terminated near the VM fascicles. When mutant for cut the axons of these cells (red) instead terminated near the DM fascicles. Fas2+ tracts are shown in magenta. Scale bars: 50 µm in B-F′; 10 µm in G,H.

Axon morphology and position in the neuropil are also distinguishing features of sensory cell types. Cut+ touch-sensing and nociceptive neurons (classes II, III and IV) terminate in the ventral neuropil, whereas Cut proprioceptive neurons (class I and dbd) terminate more dorsally (Merritt and Whitington, 1995; Schrader and Merritt, 2000; Grueber et al., 2007). We assessed the positions of afferent terminals relative to Fasciclin 2 (Fas2)-labeled ventromedial (VM) or dorsomedial (DM) fascicles. In contrast to the targeting of wild-type neurons to the VM fascicle, the terminals of a subset of cut mutant MARCM clones were repositioned to the DM fascicle (n=20; Fig. 1G-J). Notably, axon mistargeting to the DM fascicle was specific to the neurons (ddaA, ddaB, ddaF and ldaB) that also showed severe dendritic growth phenotypes upon loss of Cut (Fig. S1C,D). These data suggest that Cut is required to repress morphological characteristics of proprioceptive neurons in a subset of dorsal cluster touch-sensing neurons.

Loss of Cut leads to ectopic Pdm1/2 expression in neurons showing stunted dendritic growth

Cut has been shown to repress Pdm1 [Nubbin (Nub) – FlyBase] in embryonic sensory neurons (Brewster et al., 2001), prompting us to study this factor and the closely related transcription factor Pdm2 (collectively Pdm1/2) in more detail. Pdm1/2 are normally co-expressed in dbd and dmd1 sensory neurons, both of which are Cut and predicted to function as proprioceptors (Billin et al., 1991; Dick et al., 1991; Brewster et al., 2001; Hughes and Thomas, 2007). dbd resides along a connective tissue strand and extends dendrites in a simple bipolar fashion to span each segment (Schrader and Merritt, 2007). We found that dmd1 resides on the body wall and extends a compact dendrite bundle away from the body wall to the intersegmental nerve (ISN) (Fig. 2A,B). Thus, in contrast to da neurons, Pdm1/2+ neurons extend simple dendrites that are not associated with an epidermal substrate. dbd and dmd1 axons both terminate near the DM fascicle, consistent with a proprioceptive function, but arrive via distinct paths, with dbd axons traveling through the dorsal neuropil (Schrader and Merritt, 2000) and dmd1 axons taking a ventral-to-dorsal route (Fig. 2C,D). Thus, the normal dendritic and axonal projections of Pdm1/2-expressing dbd and dmd1 resembled those of the transformed cut clones described above.

Fig. 2.

Morphology of wild-type Pdm1/2-expressing cells and cell-autonomous suppression of Pdm1/2 by Cut in dorsal cluster neurons. (A) Wild-type morphology of dmd1 and dbd in third instar larvae as shown by labeling with mCD8::GFP driven by pdm1-Gal4 (green). Anti-HRP (magenta) labels all neuronal membranes. (Aʹ) GFP channel only. Arrow indicates axon. Dorsal is to the top and anterior is to the left in all panels. (B) Orthogonal view shows dmd1 dendrites (arrowhead) extending away from the epidermis to the intersegmental nerve (ISN). (Bʹ) GFP channel only. (C) Axon projection of wild-type dmd1 MARCM clone extends to the VNC medial fascicle (arrow; fascicles labeled in magenta). (D,Dʹ) Orthogonal views at the positions indicated by the dashed lines in C show termination near the DM fascicle (D, arrow) and the trajectory through the neuropil (Dʹ, dashed arrow). (E) Cut and Pdm2 immunoreactivity in embryonic md neurons (labeled by the E7-2-36-lacZ enhancer trap). In cut heterozygotes, Pdm2 is limited to dmd1 and dbd. (F) Pdm2 immunoreactivity is expanded to additional dorsal da neurons in cut homozygous mutant embryos. (G) Control ddaA MARCM clone labeled with anti-GFP (green) lacks Pdm2 expression (magenta). Inset is a magnified view of the Pdm2 channel. (H) cutc145 ddaA MARCM clone shows ectopic Pdm2 expression. Inset is a magnified view of the Pdm2 channel. (I) Wild-type ldaB (class III) clone does not show immunoreactivity for Pdm2 (inset). (J) cutc145 ldaB clone showing reduced terminal branching but intact primary scaffold does not show immunoreactivity for Pdm2 (inset). (K) cutc145 ldaB clone with a stunted primary outgrowth shows immunoreactivity for Pdm2 (inset). (L) Quantification of Pdm1/2 expression in cutc145 clones reveals a correlation between dendritic transformation and the misregulation of Pdm1/2. (M) Wild-type dorsal cluster. Md sensory neurons are labeled with 109(2)80-Gal4, UAS-mCD8::GFP (green) and anti-Pdm1 antibody (magenta). (M′) Anti-Pdm1-labeling is seen exclusively in the dmd1 and dbd neurons. (N) Dorsal cluster in which UAS-cut and UAS-mCD8::GFP are driven by 109(2)80-Gal4. (N′) Absence of anti-Pdm1 labeling in dmd1 and dbd when UAS-cut is driven in these cells. (O) Wild-type morphology of dmd1 FLP-out clone. (P) dmd1 FLP-out clone expressing UAS-cut shows dendritic overgrowth. Arrows indicate dendritic overgrowth along the epidermis. Scale bars: 50 µm in A,G-K,M-P; 10 µm in C-F.

Fig. 2.

Morphology of wild-type Pdm1/2-expressing cells and cell-autonomous suppression of Pdm1/2 by Cut in dorsal cluster neurons. (A) Wild-type morphology of dmd1 and dbd in third instar larvae as shown by labeling with mCD8::GFP driven by pdm1-Gal4 (green). Anti-HRP (magenta) labels all neuronal membranes. (Aʹ) GFP channel only. Arrow indicates axon. Dorsal is to the top and anterior is to the left in all panels. (B) Orthogonal view shows dmd1 dendrites (arrowhead) extending away from the epidermis to the intersegmental nerve (ISN). (Bʹ) GFP channel only. (C) Axon projection of wild-type dmd1 MARCM clone extends to the VNC medial fascicle (arrow; fascicles labeled in magenta). (D,Dʹ) Orthogonal views at the positions indicated by the dashed lines in C show termination near the DM fascicle (D, arrow) and the trajectory through the neuropil (Dʹ, dashed arrow). (E) Cut and Pdm2 immunoreactivity in embryonic md neurons (labeled by the E7-2-36-lacZ enhancer trap). In cut heterozygotes, Pdm2 is limited to dmd1 and dbd. (F) Pdm2 immunoreactivity is expanded to additional dorsal da neurons in cut homozygous mutant embryos. (G) Control ddaA MARCM clone labeled with anti-GFP (green) lacks Pdm2 expression (magenta). Inset is a magnified view of the Pdm2 channel. (H) cutc145 ddaA MARCM clone shows ectopic Pdm2 expression. Inset is a magnified view of the Pdm2 channel. (I) Wild-type ldaB (class III) clone does not show immunoreactivity for Pdm2 (inset). (J) cutc145 ldaB clone showing reduced terminal branching but intact primary scaffold does not show immunoreactivity for Pdm2 (inset). (K) cutc145 ldaB clone with a stunted primary outgrowth shows immunoreactivity for Pdm2 (inset). (L) Quantification of Pdm1/2 expression in cutc145 clones reveals a correlation between dendritic transformation and the misregulation of Pdm1/2. (M) Wild-type dorsal cluster. Md sensory neurons are labeled with 109(2)80-Gal4, UAS-mCD8::GFP (green) and anti-Pdm1 antibody (magenta). (M′) Anti-Pdm1-labeling is seen exclusively in the dmd1 and dbd neurons. (N) Dorsal cluster in which UAS-cut and UAS-mCD8::GFP are driven by 109(2)80-Gal4. (N′) Absence of anti-Pdm1 labeling in dmd1 and dbd when UAS-cut is driven in these cells. (O) Wild-type morphology of dmd1 FLP-out clone. (P) dmd1 FLP-out clone expressing UAS-cut shows dendritic overgrowth. Arrows indicate dendritic overgrowth along the epidermis. Scale bars: 50 µm in A,G-K,M-P; 10 µm in C-F.

Using antibodies against Pdm1 and Pdm2, we confirmed that Pdm1/2 expression was expanded in cut mutant embryos to include extra da neurons, which we assigned by position as ddaA, ddaB and ddaF (Fig. 2E,F). To determine whether Cut represses Pdm1/2 expression cell-autonomously we assessed Pdm1/2 expression in cut MARCM clones. We found strong expression of Pdm1/2 in cutc145 clones of ddaA, ddaB and ddaF (Fig. 2G,H,L, Fig. S1E) but not in class IV or class I neurons (Fig. S1F-H). Thus, ectopic Pdm1/2 expression was observed only in neurons that showed dendritic or axonal transformations. The strong correlation is exemplified by phenotypes observed for the lateral class III ldaB neuron. For this cell, the dendritic transformation phenotype was observed in only a subset of MARCM clones (transformation n=12/15), and ectopic expression of Pdm1/2 was observed only in transformed clones (Fig. 2I-L). Ectopic expression of Cut was sufficient to suppress Pdm1/2 expression (Brewster et al., 2001) and promote ectopic dendritic branching in dmd1 and dbd neurons (Fig. 2M-P). Together, these data point to a role for Cut in suppressing Pdm1/2 expression in dorsal/lateral class II and III neurons.

Loss-of-function evidence that Pdm1/2 suppress dendritic growth

We next examined the role of Pdm1/2 in specifying dbd and dmd1 morphology. Mutation of either pdm1 or pdm2 individually caused no morphological defects and thus each is separately dispensable for dendritogenesis (Fig. S2A,B), consistent with similar redundant function in CNS neuroblasts (Yeo et al., 1995; Grosskortenhaus et al., 2006). We characterized a lethal allele of pdm1, nubR5, generated by imprecise P-element excision (Terriente et al., 2008), and found that homozygous embryos and MARCM clones lacked detectable expression of both Pdm1 and Pdm2, indicating that this deletion is likely to be a Pdm1/2 protein null (Fig. S2C). MARCM analysis revealed a dendritic overgrowth phenotype in nubR5 dmd1 and dbd clones. Most dmd1 clones projected dendrites across the epidermis rather than the ISN (Fig. 3A-D). Most dbd clones retained their longitudinal dendrites, but sprouted ectopic branches that grew along the epidermis (Fig. 3E-H). We did not observe Cut immunoreactivity in dmd1 or dbd nubR5 clones, indicating that Pdm1/2 does not reciprocally repress Cut (Fig. S2D,E). ddaC nubR5 clones (class IV, normally Pdm1/2) showed reduced growth (n=8; Fig. S2F,G), although we noted no obvious defects in class III arborization patterns (n=3). Thus, one or more genes within the nubR5 deficiency might promote complex dendritic branching in class IV da neurons.

Fig. 3.

Genetic deletion of the pdm1/2 region leads to dendritic overgrowth. (A) Wild-type dmd1 MARCM clone. Dendrites project to the ISN (yellow arrow). Arrowhead in this and subsequent panels indicates the axon. (B) Dendrites of nubR5 dmd1 MARCM clone arborize on the epidermis (white arrows). (C) Tracings of a wild-type (boxed, top left) and mutant nubR5 dmd1 MARCM clones. Red arrow indicates normal dendritic pattern and black arrows indicate ectopic growth along the epidermis. (D) Classification of nubR5 dmd1 dendrite phenotypes (see Materials and Methods for classification criteria). (E) Wild-type dbd MARCM clone showing simple bipolar morphology (yellow arrows). (F) Aberrant growth and branching of dendrites (white arrows) in a nubR5 dbd MARCM clone. Bipolar dendrites are still seen (yellow arrows). (G) Tracings of a wild-type (top) and nubR5 dbd clones. Red arrows indicate typical branches and black arrows indicate extra branches. (H) Classification of nubR5 dbd dendrite phenotypes (see Materials and Methods for classification criteria). (I) Control dorsal cluster from a nubR5/+ first instar larvae. Animals were live imaged using clh8-Gal4, UAS-mCD8::GFP. Arrows indicate the normal bipolar morphology of dbd. Asterisks mark cell bodies of other labeled neurons. (J) Tracing of cells in panel I to illustrate normal morphology. dbd displays a characteristic bipolar morphology in control animals. (K,L) Examples of dbd dendrite overgrowth in nubR5/Df(2L) ED773 heteroallelic animals. Abnormalities in dbd morphology included branching of longitudinal dendrites (white arrows) and growth of dorsally extending arborized dendrites (red arrow). Scale bars: 50 µm in A-C,E-G; 25 µm in I-L.

Fig. 3.

Genetic deletion of the pdm1/2 region leads to dendritic overgrowth. (A) Wild-type dmd1 MARCM clone. Dendrites project to the ISN (yellow arrow). Arrowhead in this and subsequent panels indicates the axon. (B) Dendrites of nubR5 dmd1 MARCM clone arborize on the epidermis (white arrows). (C) Tracings of a wild-type (boxed, top left) and mutant nubR5 dmd1 MARCM clones. Red arrow indicates normal dendritic pattern and black arrows indicate ectopic growth along the epidermis. (D) Classification of nubR5 dmd1 dendrite phenotypes (see Materials and Methods for classification criteria). (E) Wild-type dbd MARCM clone showing simple bipolar morphology (yellow arrows). (F) Aberrant growth and branching of dendrites (white arrows) in a nubR5 dbd MARCM clone. Bipolar dendrites are still seen (yellow arrows). (G) Tracings of a wild-type (top) and nubR5 dbd clones. Red arrows indicate typical branches and black arrows indicate extra branches. (H) Classification of nubR5 dbd dendrite phenotypes (see Materials and Methods for classification criteria). (I) Control dorsal cluster from a nubR5/+ first instar larvae. Animals were live imaged using clh8-Gal4, UAS-mCD8::GFP. Arrows indicate the normal bipolar morphology of dbd. Asterisks mark cell bodies of other labeled neurons. (J) Tracing of cells in panel I to illustrate normal morphology. dbd displays a characteristic bipolar morphology in control animals. (K,L) Examples of dbd dendrite overgrowth in nubR5/Df(2L) ED773 heteroallelic animals. Abnormalities in dbd morphology included branching of longitudinal dendrites (white arrows) and growth of dorsally extending arborized dendrites (red arrow). Scale bars: 50 µm in A-C,E-G; 25 µm in I-L.

To narrow the region of the nubR5 deficiency responsible for the overgrowth phenotypes we analyzed dbd morphology in nubR5/Df(2L)ED773 animals. This combination of deficiencies eliminates Pdm1 and Pdm2 expression and putatively disrupts only two other genes in the region, RNA and export factor binding protein 2 (Ref2; coding region deleted) and bunched (bun; loss of all upstream sequence and part of its 5′ sequence). We visualized dbd morphology using live imaging in surviving early first instar larvae labeled with clh8-Gal4, UAS-mCD8::GFP and observed abnormal morphologies with excessive dendritic growth and branching (n=6; Fig. 3I-L, Fig. S2H), consistent with the overgrowth phenotypes seen in dbd nubR5 MARCM clones.

In contrast to the overgrowth and targeting defects observed in dendrites, axons in nubR5 dmd1 and dbd MARCM clones had grossly normal morphologies and retained their terminations near the DM fascicle (Fig. S2I-K). These loss-of-function studies, together with misexpression experiments presented below, point to a role for Pdm1/2 in restricting dendritic growth, but not in directing axonal targeting, of dmd1 and dbd neurons.

Ectopic Pdm1/2 expression restricts dendritic growth

To test whether Pdm1/2 expression is sufficient to restrict dendritic growth we drove expression of each gene with Gal4-109(2)80 and resolved dendritic morphologies using the FLP-out method (Struhl and Basler, 1993). Expression of either gene in class II, III or IV neurons dramatically reduced dendritic growth and branching (Fig. 4, Fig. S3). Misexpression did not cause targeting of dendrites to the ISN or to connective tissue, as seen in wild-type Pdm1/2+ neurons. Thus, although Pdm1/2 can strongly suppress growth and branching, Pdm1/2 misexpression is not able to shift dendrite substrate preference or terminal targeting. Growth and targeting might be under separate transcriptional control in Pdm1/2-expressing neurons, or the timing of Pdm1/2 expression might be critical for dendritic targeting.

Fig. 4.

Overexpression of Pdm1 or Pdm2 inhibits dendritic growth and branching in class III neurons. (A) Wild-type class III neuron ddaA, visualized using the FLP-out approach with 109(2)80-Gal4, UAS-FRT-rCD2-FRT-mCD8::GFP. (B,C) Class III ddaA neuron FLP-out clones expressing (B) UAS-pdm1 or (C) UAS-pdm2. (D,E) Total dendrite length (D) and total branch point number (E) were significantly reduced upon Pdm1 (***P<0.0001) or Pdm2 (***P<0.0001) overexpression. (F) Branch points per total dendrite length (µm) was significantly reduced upon Pdm1 (**P=0.003) but not Pdm2 (P=0.0838) overexpression. n.s., not statistically significant. Scale bars: 50 µm.

Fig. 4.

Overexpression of Pdm1 or Pdm2 inhibits dendritic growth and branching in class III neurons. (A) Wild-type class III neuron ddaA, visualized using the FLP-out approach with 109(2)80-Gal4, UAS-FRT-rCD2-FRT-mCD8::GFP. (B,C) Class III ddaA neuron FLP-out clones expressing (B) UAS-pdm1 or (C) UAS-pdm2. (D,E) Total dendrite length (D) and total branch point number (E) were significantly reduced upon Pdm1 (***P<0.0001) or Pdm2 (***P<0.0001) overexpression. (F) Branch points per total dendrite length (µm) was significantly reduced upon Pdm1 (**P=0.003) but not Pdm2 (P=0.0838) overexpression. n.s., not statistically significant. Scale bars: 50 µm.

Scalloped is expressed in a subset of da neurons

The above data demonstrate a role for Cut in directing dorsal cluster class II and III da neurons toward a complex dendritic morphology via suppression of a default Pdm1/2-dependent program of proprioceptor morphogenesis. Diversification of class II and class III sensory terminal dendrites depends at least in part on differential Cut expression levels, with high levels in class III neurons and low levels in class II neurons (Grueber et al., 2003). We explored the basis of differential Cut expression as a route to further dendritic diversification. From a collection of GFP trap lines (Morin et al., 2001; Buszczak et al., 2007; Quinones-Coello et al., 2007) we identified prominent expression of the TEAD/TEF-1 transcription factor Scalloped (Sd) (Campbell et al., 1992) in the class II neuron ddaB and the class III neuron ddaF in third instar larvae (Fig. 5A). Very low GFP expression could occasionally be observed in the class I neuron ddaE and in the class III neuron ddaA. We used an antibody against Sd that had previously revealed its expression in sensory neurons of the embryonic PNS (Guss et al., 2013), and confirmed Sd expression in ddaB and ddaF neurons in third instar larvae consistent with the GFP trap (Fig. 5B). Expression of Sd in a subset of Cut+ da neurons and the interaction between Sd and Cut in wing development (Morcillo et al., 1996) led us to examine a possible role in dendritic diversification.

Fig. 5.

Sd is required in ddaB to repress a class III-like morphology. (A) Labeling of third instar larvae with anti-HRP (red), anti-GFP (green) and anti-Cut (blue). Sd::GFP expression is strong in the class II neuron ddaB and the class III neuron ddaF in larval stages (arrows). (B) Labeling of third instar larvae with anti-Cut (green), anti-Sd (red) and anti-Elav (blue). Anti-Sd labeling is seen in the class II neuron ddaB and class III neuron ddaF (arrows). (C) Dendrites of wild-type ddaB MARCM clone (arrowheads). (D) sdΔB ddaB MARCM clone showing extensive terminal branching. (E) Enlarged views of the arborization patterns of wild-type ddaB (from C), sdΔB ddaB (from D), and wild-type ddaA MARCM clones. (F) Quantification of branch points per total dendrite length. sdΔB ddaA (P>0.9999) and sdΔB ddaF (P=0.0714) were not significantly different from wild-type clones. sdΔB ddaB clones showed significant (***P<0.0001) increases in branches per length, rendering them statistically indistinguishable from ddaA on this measure. (G) Quantification of total branch points for wild-type and sdΔB ddaA (P=0.9555), ddaB (***P<0.0001) and ddaF (*P=0.0224) clones. Scale bars: 25 μm in A,B,E; 50 μm in C,D.

Fig. 5.

Sd is required in ddaB to repress a class III-like morphology. (A) Labeling of third instar larvae with anti-HRP (red), anti-GFP (green) and anti-Cut (blue). Sd::GFP expression is strong in the class II neuron ddaB and the class III neuron ddaF in larval stages (arrows). (B) Labeling of third instar larvae with anti-Cut (green), anti-Sd (red) and anti-Elav (blue). Anti-Sd labeling is seen in the class II neuron ddaB and class III neuron ddaF (arrows). (C) Dendrites of wild-type ddaB MARCM clone (arrowheads). (D) sdΔB ddaB MARCM clone showing extensive terminal branching. (E) Enlarged views of the arborization patterns of wild-type ddaB (from C), sdΔB ddaB (from D), and wild-type ddaA MARCM clones. (F) Quantification of branch points per total dendrite length. sdΔB ddaA (P>0.9999) and sdΔB ddaF (P=0.0714) were not significantly different from wild-type clones. sdΔB ddaB clones showed significant (***P<0.0001) increases in branches per length, rendering them statistically indistinguishable from ddaA on this measure. (G) Quantification of total branch points for wild-type and sdΔB ddaA (P=0.9555), ddaB (***P<0.0001) and ddaF (*P=0.0224) clones. Scale bars: 25 μm in A,B,E; 50 μm in C,D.

Sd suppresses a class III-like terminal branching pattern in ddaB

To examine whether Sd regulates sensory neuron morphology, we generated homozygous mutant sd MARCM clones. We did not observe any obvious phenotypes for class I or IV neurons. We found no clones with a typical ddaB morphology (0/90). By contrast, among wild-type clones ddaB made up 12% (14/119) of the total. These data suggested that Sd could be required for either the production, survival or proper differentiation of ddaB. Using the md-neuron marker E7-2-36-lacZ, we found no difference in the number of md neurons in the dorsal cluster in sdΔB embryos [8.0±0.0 cells (±s.d.), n=53 clusters] compared with the sd+/− control (8.0±0.24 cells, n=6 clusters). These data argue that Sd is not required for the survival or initial generation of ddaB, and raised the possibility of a role in morphogenesis.

We investigated whether sd mutant ddaB clones show morphological transformations. Examination of sdΔB clones revealed mutant neurons in the typical location of ddaB that had developed a highly branched morphology with the short fine terminal dendrite ‘spikes’ that are a hallmark of class III neurons. We inferred the identity of these clones by verifying the presence of ddaA as well as the absence of the ddaB-characteristic dendritic arbor using HRP labeling (Fig. S4A,B). Quantification of the dendrites of these clones revealed they were statistically indistinguishable from wild-type class III ddaA clones in each of the parameters examined (Fig. 5C-G, Fig. S4E). Relative to ddaB wild-type clones, sd clones showed dramatically increased terminal branching (Fig. 5F,G). Similar dendritic terminal phenotypes were observed with the sdΔC allele (n=3). A modest, but significant increase in total dendritic branch points was also observed for ddaF, which also expresses Sd, without a change in main arbor length (Fig. 5F,G). We did not observe transformations of other class II neurons to a class III-like morphology (ldaA, n=8; vdaA, n=4; Fig. S4C,D).

We conclude that Sd is required in ddaB to repress elaboration of a class III-like morphology and in ddaF to limit terminal branching. Our data also suggest that different genetic programs specify class II neuron morphology in different regions of the body wall.

Sd and Vestigial suppress Cut expression to low levels

One hallmark of class II versus class III identity is the expression level of Cut. Class II neurons express low levels of Cut, whereas class III neurons express high levels that promote extensive terminal branching. To examine whether sd class II ddaB clones with altered morphology also show altered Cut expression we quantified the levels of Cut immunoreactivity in sd−/− ddaB clones relative to adjacent sd+/− class III ddaA neurons. As controls, we quantified Cut levels in wild-type ddaB clones relative to their neighboring ddaA neurons, as well as the levels of Cut in ddaB and ddaA neurons in sd+/− larvae. In both controls, levels of Cut immunoreactivity in ddaB neurons were, on average, about half those of ddaA neurons (mean CutddaB:CutddaA ratio=0.41; Fig. 6A,I). By contrast, sd mutant ddaB clones showed high levels of Cut that were indistinguishable from those of ddaA (mean CutddaB:CutddaA ratio=0.98; Fig. 6B,I). These results suggest that Sd is required in ddaB to limit the Cut expression level.

Fig. 6.

Sd and Vg are required for low Cut expression levels in ddaB. (A) Anti-HRP labeling of control dorsal cluster. Individual da neurons are named. (A′) Anti-Cut labeling of dorsal cluster neurons in grayscale (left) and pseudo-color image (look-up table, LUT; right). ddaA (arrowhead) and ddaB (arrow) neurons are outlined by a green box. (B) Anti-HRP labeling of dorsal cluster. ddaB neuron is mutant for sd. (B′) Anti-Cut labeling of dorsal cluster neurons in grayscale (left) and pseudo-color image (right). ddaA (arrowhead) and ddaB (arrow) neurons are outlined by a green box. (C) Wild-type MARCM clone of ddaB (arrow) showing location of cell body relative to the class III neuron ddaA (arrowhead). Clone is labeled by anti-GFP and neurons are labeled by anti-HRP. (D) Tracing of clone showing wild-type ddaB neuron (green, arrow) and terminal extensions of class III neurons (magenta, arrowhead). (E) Anti-Cut labeling of wild-type clone in C showing immunoreactivity in ddaB (arrow) and ddaA (arrowhead). Lower panel shows pseudo-coloring of anti-Cut label. (F) vg ddaB MARCM clone (arrow) shows a class III-like branching morphology. (F′) Arbors of vg clone show numerous fine terminal processes. (G) Tracing of vg clone (green, arrow) and the class III neuron ddaA (magenta, arrowhead). (H) Cut labeling of vg ddaB MARCM clone (arrow) and ddaA (arrowhead). Lower panel shows pseudo-coloring of anti-Cut label. (I) Quantification of Cut labeling in wild-type and sd ddaB neurons. Ratios of ddaB:ddaA Cut labeling in control genotypes (sd+/+ ddaB:sd+/+ ddaA, and sd+/ ddaB:sd+/ ddaA) were statistically equivalent. The ratio of ddaB:ddaA Cut labeling between sd/ ddaB clones and sd+/ ddaA was significantly increased compared with both wild-type controls (**P=0.0017) and heterozygous controls (***P=0.0004). Significance was determined by Kruskal–Wallis with Dunn's multiple comparisons test. (J) Quantification of Cut labeling in wild-type and vg ddaB neurons. The ratio of ddaB:ddaA Cut labeling is significantly increased for vg ddaB clones (***P=0.0006). Scale bars: 50 μm in A-D,F,G; 10 μm in E,H.

Fig. 6.

Sd and Vg are required for low Cut expression levels in ddaB. (A) Anti-HRP labeling of control dorsal cluster. Individual da neurons are named. (A′) Anti-Cut labeling of dorsal cluster neurons in grayscale (left) and pseudo-color image (look-up table, LUT; right). ddaA (arrowhead) and ddaB (arrow) neurons are outlined by a green box. (B) Anti-HRP labeling of dorsal cluster. ddaB neuron is mutant for sd. (B′) Anti-Cut labeling of dorsal cluster neurons in grayscale (left) and pseudo-color image (right). ddaA (arrowhead) and ddaB (arrow) neurons are outlined by a green box. (C) Wild-type MARCM clone of ddaB (arrow) showing location of cell body relative to the class III neuron ddaA (arrowhead). Clone is labeled by anti-GFP and neurons are labeled by anti-HRP. (D) Tracing of clone showing wild-type ddaB neuron (green, arrow) and terminal extensions of class III neurons (magenta, arrowhead). (E) Anti-Cut labeling of wild-type clone in C showing immunoreactivity in ddaB (arrow) and ddaA (arrowhead). Lower panel shows pseudo-coloring of anti-Cut label. (F) vg ddaB MARCM clone (arrow) shows a class III-like branching morphology. (F′) Arbors of vg clone show numerous fine terminal processes. (G) Tracing of vg clone (green, arrow) and the class III neuron ddaA (magenta, arrowhead). (H) Cut labeling of vg ddaB MARCM clone (arrow) and ddaA (arrowhead). Lower panel shows pseudo-coloring of anti-Cut label. (I) Quantification of Cut labeling in wild-type and sd ddaB neurons. Ratios of ddaB:ddaA Cut labeling in control genotypes (sd+/+ ddaB:sd+/+ ddaA, and sd+/ ddaB:sd+/ ddaA) were statistically equivalent. The ratio of ddaB:ddaA Cut labeling between sd/ ddaB clones and sd+/ ddaA was significantly increased compared with both wild-type controls (**P=0.0017) and heterozygous controls (***P=0.0004). Significance was determined by Kruskal–Wallis with Dunn's multiple comparisons test. (J) Quantification of Cut labeling in wild-type and vg ddaB neurons. The ratio of ddaB:ddaA Cut labeling is significantly increased for vg ddaB clones (***P=0.0006). Scale bars: 50 μm in A-D,F,G; 10 μm in E,H.

Either of two transcriptional intermediary factors, Vestigial (Vg) or Yorkie (Yki), interact with Sd in different tissues (Halder et al., 1998; Paumard-Rigal et al., 1998; Simmonds et al., 1998; Goulev et al., 2008; Wu et al., 2008; Zhang et al., 2008). yki MARCM clones showed normal ddaB dendritic morphology (n=6; Fig. S4F). By contrast, vg ddaB clones showed extra terminal branching and short fine terminal processes similar to class III neurons and sd mutant ddaB clones (n=5; Fig. 6C,D,F-G). As with sd mutants, Cut immunoreactivity in vg ddaB clones was significantly increased (Fig. 6E,H,J). In addition, we found that a Vg::mCitrine fusion protein is expressed in ddaB and ddaF and that anti-Vg immunolabeling overlapped with Sd::GFP expression in those neurons (Fig. S4G,H). These results suggest that Sd and Vg function together in ddaB to repress class III characteristics, including a high Cut expression level and extensive terminal branching.

Sd is sufficient to suppress terminal branching in dorsal class III neurons

To examine whether Sd is sufficient to suppress branching in high Cut-expressing class III neurons we performed ectopic expression experiments. We examined the consequences of ectopic sd expression under the control of the tubulin (αTub84B) promoter (tub>HA-sd). tub>HA-sd rescues lethality and wing phenotypes of sd mutants and thus can functionally substitute for sd. Furthermore, in an sd+ background tub>HA-sd does not cause lethality or generate the overexpression phenotypes caused by UAS-based overexpression, such as wing scalloping (M. Zecca, personal communication).

We again visualized individual neurons using the FLP-out technique. Class III ddaF neurons in a tub>HA-sd background showed highly simplified dendrites characterized by the loss of fine terminal branches (Fig. 7A,B,F,I), while the main arbor length of these cells was unchanged (Fig. S4I). The class III neuron ddaA also showed a significant decrease in branch points per total dendrite length and reduced branch number, whereas ddaB showed no dendritic defects (Fig. 7D,E,G,H, Fig. S4J-K). Thus, forced Sd expression can reduce dendritic branching of class III neurons. These data also suggest that either the level or the timing of endogenous Sd expression is not sufficient to strongly suppress terminal branching in ddaF. UAS-cut expression provided significant rescue of ddaF branch points per length in a tub>HA-sd background but did not restore arbors to wild-type morphology and did not restore total branch number in this cell (Fig. 7C,F,I). Altogether, the partial restoration of dendritic complexity in tub>HA-sd by UAS-cut, together with our loss-of-function data, suggest that Sd regulates dendritic morphology in part by specifying low levels of Cut expression. These data also raise the possibility that Sd can interfere with dendritic branching programs that operate downstream of, or in parallel to, Cut.

Fig. 7.

Sd driven by the tubulin promoter simplifies ddaF dendrites and this effect is partially reversed by overexpression of Cut. (A) Wild-type proximal arbor of a dorsal cluster class III neuron ddaF, visualized using the FLP-out approach with 109(2)80-Gal4, UAS-FRT-rCD2-FRT-mCD8::GFP. Terminal branches are indicated by arrows. (B) A ddaF arbor in tub>HA-sd larva. Terminal branches are indicated by arrows. A branch from the ddaA neuron, which is less affected than ddaF, is visible at the bottom (arrowhead). (C) A ddaF arbor when UAS-cut is expressed using 109(2)80-Gal4 together with tub>HA-sd. (D) Quantification of the effects of tub>HA-sd on branch points per total dendrite length for ddaA (*P=0.0108). (E) Quantification of effects of tub>HA-sd on branch points per total dendrite length for ddaB (P=0.278). (F) Quantification of effects of tub>HA-sd and UAS-cut on branch points per total dendrite length for ddaF. Branch points per total dendrite length was unchanged in UAS-cut compared with control (P=0.7015), but significantly decreased in tub>HA-sd compared with control (***P<0.0001). Expression of UAS-cut together with tub>HA-sd significantly increased branch density compared with tub>HA-sd alone (*P=0.0126) but did not restore wild-type density (***P<0.0001). (G) Quantification of effects of tub>HA-sd on total number of branch points in ddaA (*P=0.0351). (H) Quantification of effects of tub>HA-sd on total number of branches in ddaB (P=0.8431). (I) Quantification of effects of tub>HA-sd and UAS-cut on total number of branch points in ddaF. Compared with wild-type, branch points were significantly reduced in tub>HA-sd larvae (***P<0.0001) and in tub>HA-sd, UAS-cut larvae (*P=0.0213). Expression of UAS-cut with tub>HA-sd did not significantly affect branch point number compared with tub>HA-sd alone (P>0.9999). Significance in panel I was determined by Kruskal–Wallis with Dunn's multiple comparisons test. Scale bars: 50 μm.

Fig. 7.

Sd driven by the tubulin promoter simplifies ddaF dendrites and this effect is partially reversed by overexpression of Cut. (A) Wild-type proximal arbor of a dorsal cluster class III neuron ddaF, visualized using the FLP-out approach with 109(2)80-Gal4, UAS-FRT-rCD2-FRT-mCD8::GFP. Terminal branches are indicated by arrows. (B) A ddaF arbor in tub>HA-sd larva. Terminal branches are indicated by arrows. A branch from the ddaA neuron, which is less affected than ddaF, is visible at the bottom (arrowhead). (C) A ddaF arbor when UAS-cut is expressed using 109(2)80-Gal4 together with tub>HA-sd. (D) Quantification of the effects of tub>HA-sd on branch points per total dendrite length for ddaA (*P=0.0108). (E) Quantification of effects of tub>HA-sd on branch points per total dendrite length for ddaB (P=0.278). (F) Quantification of effects of tub>HA-sd and UAS-cut on branch points per total dendrite length for ddaF. Branch points per total dendrite length was unchanged in UAS-cut compared with control (P=0.7015), but significantly decreased in tub>HA-sd compared with control (***P<0.0001). Expression of UAS-cut together with tub>HA-sd significantly increased branch density compared with tub>HA-sd alone (*P=0.0126) but did not restore wild-type density (***P<0.0001). (G) Quantification of effects of tub>HA-sd on total number of branch points in ddaA (*P=0.0351). (H) Quantification of effects of tub>HA-sd on total number of branches in ddaB (P=0.8431). (I) Quantification of effects of tub>HA-sd and UAS-cut on total number of branch points in ddaF. Compared with wild-type, branch points were significantly reduced in tub>HA-sd larvae (***P<0.0001) and in tub>HA-sd, UAS-cut larvae (*P=0.0213). Expression of UAS-cut with tub>HA-sd did not significantly affect branch point number compared with tub>HA-sd alone (P>0.9999). Significance in panel I was determined by Kruskal–Wallis with Dunn's multiple comparisons test. Scale bars: 50 μm.

We have studied how three distinct somatosensory neuron morphologies are established in Drosophila. Together, our results reveal a repressive strategy for the diversification of dendritic arbor morphology involving the transcription factors Cut, Pdm1/2, Sd and Vg (Fig. 8).

Fig. 8.

Cut, Pdm1/2, Sd and Vg interact to diversify sensory neuron morphology. (A) Summary of the proposed regulatory interactions between Cut, Pdm1/2, Sd and Vg that regulate morphogenesis of dorsal cluster neurons. Multiple neurons have a capacity to express Pdm1/2, but expression is blocked by Cut (see Fig. 2). Pdm1/2 limit dendrite growth in dbd and dmd1 (see Fig. 3). Cut promotes growth by repressing Pdm1/2 and by promoting dendritic growth and branching. At high levels, Cut promotes the development of fine terminal branches characteristic of class III da neurons (Grueber et al., 2003). Sd and Vg repress Cut to low levels in ddaB and prevent acquisition of class III features (see Figs 5 and 6). Unlike in ddaB, Cut expression remains high in the presence of Sd and Vg in ddaF neurons. These cells develop class III-like terminal branches, although Sd limits the number of terminal branches (not shown in model; see Fig. 5). In addition to acting through Cut regulation, Sd may also act in parallel to, or downstream of, Cut to restrict terminal branching (see Fig. 7). (B) The regulatory interactions depicted in A promote specific transcription factor expression patterns as determined by antibody labeling (see Figs 2, 5, Fig. S4). (C) The interactions summarized in A contribute to the mature dendritic morphology of individual sensory neurons.

Fig. 8.

Cut, Pdm1/2, Sd and Vg interact to diversify sensory neuron morphology. (A) Summary of the proposed regulatory interactions between Cut, Pdm1/2, Sd and Vg that regulate morphogenesis of dorsal cluster neurons. Multiple neurons have a capacity to express Pdm1/2, but expression is blocked by Cut (see Fig. 2). Pdm1/2 limit dendrite growth in dbd and dmd1 (see Fig. 3). Cut promotes growth by repressing Pdm1/2 and by promoting dendritic growth and branching. At high levels, Cut promotes the development of fine terminal branches characteristic of class III da neurons (Grueber et al., 2003). Sd and Vg repress Cut to low levels in ddaB and prevent acquisition of class III features (see Figs 5 and 6). Unlike in ddaB, Cut expression remains high in the presence of Sd and Vg in ddaF neurons. These cells develop class III-like terminal branches, although Sd limits the number of terminal branches (not shown in model; see Fig. 5). In addition to acting through Cut regulation, Sd may also act in parallel to, or downstream of, Cut to restrict terminal branching (see Fig. 7). (B) The regulatory interactions depicted in A promote specific transcription factor expression patterns as determined by antibody labeling (see Figs 2, 5, Fig. S4). (C) The interactions summarized in A contribute to the mature dendritic morphology of individual sensory neurons.

Repression of alternative developmental programs promotes sensory neuron morphological diversification

Cut/Cux transcription factors have conserved roles in dendritic morphogenesis, including control of dendritic elaboration of da neurons (Grueber et al., 2003; Jinushi-Nakao et al., 2007), dendritic targeting of Drosophila olfactory projection neurons (Komiyama and Luo, 2007), and control of dendritic morphogenesis of upper layer cortical neurons in vertebrates (Cubelos et al., 2010). Previous studies showing that Cut levels impact class-specific complexity of da neuron dendritic arbors (Grueber et al., 2003) raised several questions, including how Cut activity establishes divergent sensory neuron morphologies, how different levels of Cut are specified, and whether Cut controls axonal targeting as well as dendritic branching in da neurons. Our data begin to address these questions by characterizing regulatory interactions with Pdm1/2, Sd and Vg, and by identifying a role for Cut in da neuron axon targeting (Fig. 8).

Loss-of-function and gain-of-function experiments indicate that Pdm1 and Pdm2 inhibit dendritic growth. Cut represses the action of Pdm1/2 and, by extension, derepresses a program for extensive dendritic elaboration (Fig. 8). Complementing level-dependent functions for Cut, this function of Cut is likely to be level independent as both low (class II) and high (class III) levels appear sufficient to repress Pdm1/2 expression. We suggest that a minimum level of Cut is required for Pdm1/2 repression in dorsal cluster class II and III neurons, allowing for the establishment of an expansive epidermal territory. Higher levels of Cut can promote increased higher-order branching and further diversify sensory neuron morphology. POU domain transcription factors are important regulators of neuronal morphogenesis in both vertebrate and invertebrate sensory systems (Komiyama et al., 2003; Badea et al., 2009) and it would be interesting to determine whether repression of alternative or default states is a more general mechanism by which transcriptional programs involving POU proteins generate diversity in neuronal morphology.

In the fly somatosensory system, sensory neuron subtype is correlated with the morphology of dendrites in the periphery and the positioning of sensory afferents centrally. While the mechanisms that differentiate dendrite versus axon development are beginning to be uncovered (Ye et al., 2007, 2011; Satoh et al., 2008; Zheng et al., 2008), the mechanisms that coordinate md neuron dendrite and axon morphology have not been established. Our results suggest that Cut fulfills such a role in a subset of somatosensory neurons. Loss of Cut led to the development of proprioceptive dendritic features and to a shift in axon targeting to regions where proprioceptive neurons terminate. Still unknown are the mechanisms by which Cut regulates this switch, as Pdm1/2 appear not to affect axon terminal positioning of da neurons. Prior studies have shown that axon positioning along the dorsoventral axis is controlled in part by Semaphorin-Plexin signaling (Zlatic et al., 2009), and it will be important to examine whether Cut directly or indirectly regulates this pathway to control axonal targeting.

Transcriptional control of Cut levels

Different levels of transcriptional regulators can affect neuronal morphogenesis (Grueber et al., 2003; Chen et al., 2006), but how specific levels are established is not understood. We found that Sd and Vg establish low Cut levels and repress a more highly branched class III morphology in ddaB (Fig. 8). Sd and Vg are also expressed in ddaF, which develops a class III-like branching pattern, and our loss-of-function experiments suggest that Sd has a modest effect on branching in this cell as well. Our results also raise the possibility (not schematized in Fig. 8) that Sd might also act via a parallel pathway to set an upper limit on the branch-promoting activity of Cut, perhaps by blocking pathways that are required downstream of Cut for dendrogenic activity. In C. elegans sensory neuron specification, the AHR-1 transcription factor operates by controlling both a transcription factor and a subset of its downstream targets (Smith et al., 2013). Dual-level regulatory strategies might add robustness and range to transcription factor encoding of specific morphological features or identity. The role of class III terminal branches in mediating responsiveness to gentle touch (Tsubouchi et al., 2012; Bagley et al., 2014) raises the possibility that Sd regulation of morphology could fine-tune mechanoreceptor sensitivity.

Sd and Cut interact during wing development (Morcillo et al., 1996), and during adult development Vg suppresses Cut levels in a subset of muscle precursors (Sudarsan et al., 2001). Sd and Vg therefore operate in several contexts to regulate Cut levels and promote cell diversification, and our results identify roles in differentiating neurons in the control of dendrite morphology. Similarly, in C. elegans, the sd homolog egl-44 suppresses late stage ectopic branching in multidendritic PVD neurons (Smith et al., 2010). Given these roles in dendritic branching in invertebrate neurons, and the broad expression of Tead1 (the mouse homolog of Sd) in the mouse brain (Lein et al., 2007), it will be interesting to examine whether TEAD transcription factors also regulate dendritic branching in vertebrate neurons.

Possible implications for the diversification of neuronal morphology during evolution

Our findings provide an example of how linked repressive interactions among transcriptional regulators can diversify dendritic morphology. In Drosophila sensory neurons, it appears that a broad competence to produce dbd/dmd1-type morphologies in dorsal regions of the larva is restricted by the action of Cut (Fig. 8). This switch generates at least two types of sensory neurons: Cut+/Pdm (which perform tactile functions) and Cut/Pdm+ (which function as proprioceptors) (Fig. 8). Further morphological diversification of Cut+/Pdm neurons is achieved by modulation of Cut expression levels (Fig. 8). The partial, rather than complete, repression of Cut levels in ddaB becomes important in this context, since it allows for the maintenance of Pdm1/2 repression, as well as the development of a third distinct Cut-dependent morphology (Fig. 8). High-level Cut expression might represent a default state for the dorsal cluster class II neuron that is refined by Sd and Vg (Fig. 8).

Repression of alternative identities and morphologies by transcriptional regulators is an important driver of neuronal diversification during the development of sensory and motor systems (Jung et al., 2010; Li et al., 2013; Philippidou and Dasen, 2013; Gordon and Hobert, 2015). Transcription factors may suppress alternative programs of differentiation and promote neuronal diversity by direct repression of other transcription factors, repression of their downstream target genes, or by competitive binding of common co-factors to block transcription factor function (Smith et al., 2013; Borromeo et al., 2014; Gordon and Hobert, 2015). Differential expression levels of the same transcription factor can also drive distinct morphology, identity and connectivity to further increase diversity in the nervous system (Grueber et al., 2003; Chen et al., 2006; Dasen et al., 2008; Smith et al., 2013). It has been proposed that such transformations from one cell type, or cell characteristic, to another by mutations in terminal selector genes might provide a basis for the evolutionary diversification of cell types in the nervous system (Arlotta and Hobert, 2015). One speculative scenario is that, in the Drosophila PNS, the suppression of default programs in subsets of equivalent neurons by the action of transcriptional regulators provided a mechanism for the evolutionary diversification of somatosensory cell types without disruption of crucial existing modalities.

Fly stocks

Animals were reared using standard methods. We used pdm1-Gal4 [nub-Gal4 (Calleja et al., 2000)], clh8-Gal4 (Hughes and Thomas, 2007), GMR11F05-Gal4 (Pfeiffer et al., 2008; Li et al., 2014), 109(2)80-Gal4 (Gao et al., 1999) and clh201-Gal4 (Hughes and Thomas, 2007). tubP>HA-sd and FRT42D vg83b27R were provided by Drs M. Zecca and G. Struhl (Columbia University), vg::mCitrine by Dr G. Struhl (unpublished), and sdΔB and sdΔC alleles were from Dr J. Jiang (UT Southwestern) (Zhang et al., 2008). FRT42D ykiB5 was from Dr Duojia Pan (Johns Hopkins). cutc145 and cutdb3 are null alleles (Jack, 1985; Blochlinger et al., 1988). cutdb3 was combined with the md neuron marker E7-2-36 lacZ for studies of embryonic stages. The nubR5 deletion is the result of P-element excision (Terriente et al., 2008) and was provided by Dr F. Diaz-Benjumea (Universidad Autónoma de Madrid, Spain). PCR mapping revealed that the nubR5 deletion removes ∼224 kb of genomic material spanning 11 protein-coding regions, which is about half the number of genes deleted in Df(2L)ED773, which also deletes both pdm1 and pdm2 (Ryder et al., 2004, 2007; Grosskortenhaus et al., 2006). Other stocks were Df(2L)ED773 (Bloomington Stock Center), pdm2E46 (Bloomington Stock Center), Sd::GFP (CA07575; Buszczak et al., 2007), full-length UAS-cut (Ludlow et al., 1996; Grueber et al., 2003), UAS-pdm1 (Neumann and Cohen, 1998), UAS-pdm2 (Grosskortenhaus et al., 2006), UAS-mCD8::GFP and UAS-mCD8::Cherry (Bloomington Stock Center). Animals were analyzed as third instar larvae, except when noted (i.e. embryos or first instar).

Mosaic analysis

For MARCM experiments (Lee and Luo, 1999) the following fly stocks were used: (1) cutc145 FRT19A/FM7, (2) sdΔB FRT19A, (3) FRT19A;;ry, (4) tub-Gal80, hs-FLP, FRT19A; 109(2)80-Gal4, UAS-mcD8::GFP, (5) ywhs-FLP122; nubR5 FRT40A/CyO, (6) nub1 FRT40A/CyO, (7) FRT40A/CyO, (8) hs-FLP, elav-Gal4, UAS-mCD8::GFP; tub-Gal80 FRT 40A, (9) FRT42D ykiB5, (10) FRT42D vg83b27R, (11) w; FRT 42D, (12) hs-FLP, elav-Gal4, UAS-mCD8::GFP; FRT 42D tub-Gal80, (13) tub-Gal80 FRT 40A/CyO; GMR11F05-Gal4, UAS-mCD8::GFP, (14) ywhs-FLP122; FRT 40A/CyO. MARCM clones were generated as previously described (Matthews et al., 2007). For overexpression experiments, we crossed hs-FLP; 109(2)80-Gal4; UAS>rCD2>mCD8::GFP to either w1118, w1118; UAS-pdm1/CyO, Dfd-YFP, w1118; UAS-pdm2/CyO, Dfd-YFP or tub>HA-sd. FLP-out clones were generated using a 30 min heat shock at 38°C to induce mosaic expression of rCD2 and mCD8::GFP.

Immunohistochemistry

Immunohistochemistry was performed according to published methods (Grueber et al., 2002). Antibodies used were rabbit anti-Vg (1:20; Halder et al., 1998; courtesy of S. Carroll, University of Wisconsin), guinea pig anti-Sd (1:500; Guss et al., 2013), guinea pig anti-Vg (1:10; Zecca and Struhl, 2010; courtesy of G. Struhl), mouse anti-Cut (1:20; DSHB #2B10), goat anti-HRP (1:200; Jackson ImmunoResearch #123-005-021), chicken anti-GFP (1:1000; Abcam ab13970), rabbit anti-β-gal (1:200; Cappell #55976), rat anti-ELAV (1:10, DSHB #7E8A10, mouse anti-Fas2 (1:10; DSHB #1D4); rabbit anti-Pdm1 (1:1000; Yeo, 1995), and rat anti-Pdm2 (1:10; Grosskortenhaus et al., 2006). Secondary antibodies (Jackson ImmunoResearch) were used at 1:200. Following antibody labeling, animals were dehydrated in an ethanol series, cleared in xylenes, and mounted in DPX (Electron Microscopy Sciences) as described (Grueber et al., 2002). For the experiments depicted in Fig. S2I-K, animals were mounted in Vectashield (Vector Laboratories).

Image acquisition and analysis

Imaging was performed on a Zeiss 510 Meta confocal microscope with LSM software (Zeiss) using 40× Plan Neofluar and 63× objectives for all images except those in Fig. 3I-L and Fig. S2H-J, which were acquired using a 3i Everest spinning disk confocal microscope with Slidebook 6.0 (3i) imaging software and a Zeiss Plan-Apochromat 40× oil objective. Arbors were scanned as single frames or as partially overlapping frames that were assembled in Photoshop CS2 (Adobe Systems) or Neurolucida (MBF Bioscience). For quantitative analysis, neurons were traced as confocal stacks using Neurolucida and quantified using Neurolucida Explorer. Whole image adjustments of brightness and contrast were applied to aid in visualization of fine dendritic processes in figure images.

Quantification of Cut levels was performed similarly to prior studies (Grueber et al., 2003). Briefly, ddaA and ddaB Cut-labeled nuclei were identified in confocal stacks. Mean pixel intensity values for each nucleus were obtained from each successive confocal section in which the nucleus was visible using the Measure command in Fiji (Schindelin et al., 2012). The highest mean pixel value obtained for each analyzed nucleus was used to compute ddaB:ddaA ratios. Confocal settings were identical among the different imaging sessions for data collected for comparison between groups. Cut values were quantified from raw values for each image without adjustment of brightness, contrast, or levels. To aid visualization and comparison in figure panels, levels were adjusted uniformly across some images from (min-max) of 20-148 to 10-232 (Fig. 6A′) and 11-95 to 10-229 (Fig. 6B′). These changes were made following quantification.

Classification of nubR5 phenotypes: dmd1 clones were classified as ‘normal’ if all dendrites project to the ISN; ‘mild’ if some dendrites project to the ISN with some dendrites that do not grow towards the target; or ‘severe’ if there was no ISN targeting and/or substantial growth of dendrites on the epidermis. dbd clones were classified as ‘normal’ if normal unbranched bipolar morphology is maintained; ‘mild’ if there is abnormal branching or mistargeting of at least one of the main dendrites; or ‘severe’ if there is an additional, branched dorsal dendrite and/or a lack of bipolar morphology.

Statistical analysis

Statistical analysis was performed in R (R Project for Statistical Computing) and GraphPad Prism. Data were tested for normality using a Shapiro-Wilk test. Nonparametric analyses (Kruskal–Wallis with Dunn's multiple comparisons test) were used for datasets that deviated from a normal distribution as noted (Fig. S4E, Fig. 6I, Fig. 7I). For all other datasets statistical significance was determined using a Student's t-test with Welch's correction for all comparisons between two groups and one-way ANOVA with Tukey's post-hoc test for all comparisons with three or more groups. n values refer to individual cells (i.e. MARCM or FLP-out clones) except where noted in the text. No statistical methods were used to predetermine sample size. The sample sizes used are consistent with those found in similar studies of da neuron morphogenesis. Animals were assigned to groups based on genotype, and researchers were not blind to conditions. Clones were excluded from analysis if arbors were damaged during processing. Datasets presented as box and whisker plots are as follows: quartiles Q1-Q3 (25%-75%, box), median (thick line), whiskers span minimum to maximum values. All individual data points are displayed as open circles. Multiplicity-adjusted P-values are given for all ANOVA/Tukey results. In figures, *P<0.05, **P<0.01 and ***P<0.001.

We thank Dr M. Zecca for generously sharing fly stocks prior to publication; Drs J. Bell, S. Carroll, W. Chia, S. Cohen, F. Diaz-Benjumea, C. Doe, K. Guss, J. Jiang, B. Ohlstein and G. Struhl for generous sharing of fly stocks and antibodies; Dr J. Dodd for comments on the manuscript; N. Ghani for input on branching analysis; and Dr O. Hobert and members of the W.B.G. lab for discussions. M.M.C. thanks Dr M. Freeman for support during preparation of the manuscript. The anti-Cut, anti-ELAV, and anti-Fas2 antibodies were obtained from the Developmental Studies Hybridoma Bank (DSHB), created for the NICHD of the NIH and maintained at The University of Iowa, Department of Biology, Iowa City, IA 52242, USA.

Author contributions

M.M.C., J.T. and W.B.G. conceived and performed the experiments and analyzed the data. M.M.C. and W.B.G. wrote the manuscript with input from J.T.

Funding

This work was supported in part by a National Science Foundation Graduate Research Fellowship (M.M.C.), the Searle Scholars Program, an Irma T. Hirschl/Monique Weill-Caulier Trust Career Award, and the National Institutes of Health [NS061908] (to W.B.G.). Deposited in PMC for release after 12 months.

Allan
,
D. W.
and
Thor
,
S.
(
2015
).
Transcriptional selectors, masters, and combinatorial codes: regulatory principles of neural subtype specification
.
Wiley Interdiscip. Rev. Dev. Biol.
4
,
505
-
528
.
Arlotta
,
P.
and
Hobert
,
O.
(
2015
).
Homeotic transformations of neuronal cell identities
.
Trends Neurosci.
38
,
751
-
762
.
Badea
,
T. C.
,
Cahill
,
H.
,
Ecker
,
J.
,
Hattar
,
S.
and
Nathans
,
J.
(
2009
).
Distinct roles of transcription factors brn3a and brn3b in controlling the development, morphology, and function of retinal ganglion cells
.
Neuron
61
,
852
-
864
.
Bagley
,
J. A.
,
Yan
,
Z.
,
Zhang
,
W.
,
Wildonger
,
J.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2014
).
Double-bromo and extraterminal (BET) domain proteins regulate dendrite morphology and mechanosensory function
.
Genes Dev.
28
,
1940
-
1956
.
Billin
,
A. N.
,
Cockerill
,
K. A.
and
Poole
,
S. J.
(
1991
).
Isolation of a family of Drosophila POU domain genes expressed in early development
.
Mech. Dev.
34
,
75
-
84
.
Blochlinger
,
K.
,
Bodmer
,
R.
,
Jack
,
J.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
1988
).
Primary structure and expression of a product from cut, a locus involved in specifying sensory organ identity in Drosophila
.
Nature
333
,
629
-
635
.
Bodmer
,
R.
and
Jan
,
Y. N.
(
1987
).
Morphological differentiation of the embryonic peripheral neurons in Drosophila
.
Roux's Arch. Dev. Biol.
196
,
69
-
77
.
Borromeo
,
M. D.
,
Meredith
,
D. M.
,
Castro
,
D. S.
,
Chang
,
J. C.
,
Tung
,
K.-C.
,
Guillemot
,
F.
and
Johnson
,
J. E.
(
2014
).
A transcription factor network specifying inhibitory versus excitatory neurons in the dorsal spinal cord
.
Development
141
,
2803
-
2812
.
Brewster
,
R.
,
Hardiman
,
K.
,
Deo
,
M.
,
Khan
,
S.
and
Bodmer
,
R.
(
2001
).
The selector gene cut represses a neural cell fate that is specified independently of the Achaete-Scute-Complex and atonal
.
Mech. Dev.
105
,
57
-
68
.
Buszczak
,
M.
,
Paterno
,
S.
,
Lighthouse
,
D.
,
Bachman
,
J.
,
Planck
,
J.
,
Owen
,
S.
,
Skora
,
A. D.
,
Nystul
,
T. G.
,
Ohlstein
,
B.
,
Allen
,
A.
, et al. 
(
2007
).
The carnegie protein trap library: a versatile tool for Drosophila developmental studies
.
Genetics
175
,
1505
-
1531
.
Calleja
,
M.
,
Herranz
,
H.
,
Estella
,
C.
,
Casal
,
J.
,
Lawrence
,
P.
,
Simpson
,
P.
and
Morata
,
G.
(
2000
).
Generation of medial and lateral dorsal body domains by the pannier gene of Drosophila
.
Development
127
,
3971
-
3980
.
Campbell
,
S.
,
Inamdar
,
M.
,
Rodrigues
,
V.
,
Raghavan
,
V.
,
Palazzolo
,
M.
and
Chovnick
,
A.
(
1992
).
The scalloped gene encodes a novel, evolutionarily conserved transcription factor required for sensory organ differentiation in Drosophila
.
Genes Dev.
6
,
367
-
379
.
Chen
,
A. I.
,
de Nooij
,
J. C.
and
Jessell
,
T. M.
(
2006
).
Graded activity of transcription factor Runx3 specifies the laminar termination pattern of sensory axons in the developing spinal cord
.
Neuron
49
,
395
-
408
.
Corty
,
M. M.
,
Matthews
,
B. J.
and
Grueber
,
W. B.
(
2009
).
Molecules and mechanisms of dendrite development in Drosophila
.
Development
136
,
1049
-
1061
.
Crozatier
,
M.
and
Vincent
,
A.
(
2008
).
Control of multidendritic neuron differentiation in Drosophila: the role of Collier
.
Dev. Biol.
315
,
232
-
242
.
Cubelos
,
B.
,
Sebastián-Serrano
,
A.
,
Beccari
,
L.
,
Calcagnotto
,
M. E.
,
Cisneros
,
E.
,
Kim
,
S.
,
Dopazo
,
A.
,
Alvarez-Dolado
,
M.
,
Redondo
,
J. M.
,
Bovolenta
,
P.
, et al. 
(
2010
).
Cux1 and Cux2 regulate dendritic branching, spine morphology, and synapses of the upper layer neurons of the cortex
.
Neuron
66
,
523
-
535
.
Dasen
,
J. S.
and
Jessell
,
T. M.
(
2009
).
Hox networks and the origins of motor neuron diversity
.
Curr. Top. Dev. Biol.
88
,
169
-
200
.
Dasen
,
J. S.
,
De Camilli
,
A.
,
Wang
,
B.
,
Tucker
,
P. W.
and
Jessell
,
T. M.
(
2008
).
Hox repertoires for motor neuron diversity and connectivity gated by a single accessory factor, FoxP1
.
Cell
134
,
304
-
316
.
Dick
,
T.
,
Yang
,
X. H.
,
Yeo
,
S. L.
and
Chia
,
W.
(
1991
).
Two closely linked Drosophila POU domain genes are expressed in neuroblasts and sensory elements
.
Proc. Natl. Acad. Sci. USA
88
,
7645
-
7649
.
Enriquez
,
J.
,
Venkatasubramanian
,
L.
,
Baek
,
M.
,
Peterson
,
M.
,
Aghayeva
,
U.
and
Mann
,
R. S.
(
2015
).
Specification of individual adult motor neuron morphologies by combinatorial transcription factor codes
.
Neuron
86
,
955
-
970
.
Ferreira
,
T.
,
Ou
,
Y.
,
Li
,
S.
,
Giniger
,
E.
and
van Meyel
,
D. J.
(
2014
).
Dendrite architecture organized by transcriptional control of the F-actin nucleator Spire
.
Development
141
,
650
-
660
.
Gao
,
F. B.
,
Brenman
,
J. E.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
1999
).
Genes regulating dendritic outgrowth, branching, and routing in Drosophila
.
Genes Dev.
13
,
2549
-
2561
. http://genesdev.cshlp.org/content/13/19/2549
Gordon
,
P. M.
and
Hobert
,
O.
(
2015
).
A competition mechanism for a homeotic neuron identity transformation in C. elegans
.
Dev. Cell
34
,
206
-
219
.
Goulev
,
Y.
,
Fauny
,
J. D.
,
Gonzalez-Marti
,
B.
,
Flagiello
,
D.
,
Silber
,
J.
and
Zider
,
A.
(
2008
).
SCALLOPED interacts with YORKIE, the nuclear effector of the hippo tumor-suppressor pathway in Drosophila
.
Curr. Biol.
18
,
435
-
441
.
Grosskortenhaus
,
R.
,
Robinson
,
K. J.
and
Doe
,
C. Q.
(
2006
).
Pdm and Castor specify late-born motor neuron identity in the NB7-1 lineage
.
Genes Dev.
20
,
2618
-
2627
.
Grueber
,
W. B.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2002
).
Tiling of the Drosophila epidermis by multidendritic sensory neurons
.
Development
129
,
2867
-
2878
.
Grueber
,
W. B.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2003
).
Different levels of the homeodomain protein cut regulate distinct dendrite branching patterns of Drosophila multidendritic neurons
.
Cell
112
,
805
-
818
.
Grueber
,
W. B.
,
Ye
,
B.
,
Yang
,
C.-H.
,
Younger
,
S.
,
Borden
,
K.
,
Jan
,
L. Y.
and
Jan
,
Y.-N.
(
2007
).
Projections of Drosophila multidendritic neurons in the central nervous system: links with peripheral dendrite morphology
.
Development
134
,
55
-
64
.
Guss
,
K. A.
,
Benson
,
M.
,
Gubitosi
,
N.
,
Brondell
,
K.
,
Broadie
,
K.
and
Skeath
,
J. B.
(
2013
).
Expression and function of scalloped during Drosophila development
.
Dev. Dyn.
242
,
874
-
885
.
Halder
,
G.
,
Polaczyk
,
P.
,
Kraus
,
M. E.
,
Hudson
,
A.
,
Kim
,
J.
,
Laughon
,
A.
and
Carroll
,
S.
(
1998
).
The Vestigial and Scalloped proteins act together to directly regulate wing-specific gene expression in Drosophila
.
Genes Dev.
12
,
3900
-
3909
.
Hattori
,
Y.
,
Sugimura
,
K.
and
Uemura
,
T.
(
2007
).
Selective expression of Knot/Collier, a transcriptional regulator of the EBF/Olf-1 family, endows the Drosophila sensory system with neuronal class-specific elaborated dendritic patterns
.
Genes Cells
12
,
1011
-
1022
.
Hughes
,
C. L.
and
Thomas
,
J. B.
(
2007
).
A sensory feedback circuit coordinates muscle activity in Drosophila
.
Mol. Cell. Neurosci.
35
,
383
-
396
.
Hwang
,
R. Y.
,
Zhong
,
L.
,
Xu
,
Y.
,
Johnson
,
T.
,
Zhang
,
F.
,
Deisseroth
,
K.
and
Tracey
,
W. D.
(
2007
).
Nociceptive neurons protect Drosophila larvae from parasitoid wasps
.
Curr. Biol.
17
,
2105
-
2116
.
Iyer
,
S. C.
,
Ramachandran Iyer
,
E. P.
,
Meduri
,
R.
,
Rubaharan
,
M.
,
Kuntimaddi
,
A.
,
Karamsetty
,
M.
and
Cox
,
D. N.
(
2013
).
Cut, via CrebA, transcriptionally regulates the COPII secretory pathway to direct dendrite development in Drosophila
.
J. Cell Sci.
126
,
4732
-
4745
.
Jack
,
J. W.
(
1985
).
Molecular organization of the cut locus of Drosophila melanogaster
.
Cell
42
,
869
-
876
.
Jan
,
Y.-N.
and
Jan
,
L. Y.
(
2010
).
Branching out: mechanisms of dendritic arborization
.
Nat. Rev. Neurosci.
11
,
316
-
328
.
Jinushi-Nakao
,
S.
,
Arvind
,
R.
,
Amikura
,
R.
,
Kinameri
,
E.
,
Liu
,
A. W.
and
Moore
,
A. W.
(
2007
).
Knot/Collier and cut control different aspects of dendrite cytoskeleton and synergize to define final arbor shape
.
Neuron
56
,
963
-
978
.
Jung
,
H.
,
Lacombe
,
J.
,
Mazzoni
,
E. O.
,
Liem
,
K. F.
, Jr.
,
Grinstein
,
J.
,
Mahony
,
S.
,
Mukhopadhyay
,
D.
,
Gifford
,
D. K.
,
Young
,
R. A.
,
Anderson
,
K. V.
, et al. 
(
2010
).
Global control of motor neuron topography mediated by the repressive actions of a single hox gene
.
Neuron
67
,
781
-
796
.
Kim
,
M. D.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2006
).
The bHLH-PAS protein Spineless is necessary for the diversification of dendrite morphology of Drosophila dendritic arborization neurons
.
Genes Dev.
20
,
2806
-
2819
.
Komiyama
,
T.
and
Luo
,
L.
(
2007
).
Intrinsic control of precise dendritic targeting by an ensemble of transcription factors
.
Curr. Biol.
17
,
278
-
285
.
Komiyama
,
T.
,
Johnson
,
W. A.
,
Luo
,
L.
and
Jefferis
,
G. S. X. E.
(
2003
).
From lineage to wiring specificity. POU domain transcription factors control precise connections of Drosophila olfactory projection neurons
.
Cell
112
,
157
-
167
.
Landgraf
,
M.
and
Thor
,
S.
(
2006
).
Development of Drosophila motoneurons: specification and morphology
.
Semin. Cell Dev. Biol.
17
,
3
-
11
.
Lee
,
T.
and
Luo
,
L.
(
1999
).
Mosaic analysis with a repressible cell marker for studies of gene function in neuronal morphogenesis
.
Neuron
22
,
451
-
461
.
Lein
,
E. S.
,
Hawrylycz
,
M. J.
,
Ao
,
N.
,
Ayres
,
M.
,
Bensinger
,
A.
,
Bernard
,
A.
,
Boe
,
A. F.
,
Boguski
,
M. S.
,
Brockway
,
K. S.
,
Byrnes
,
E. J.
, et al. 
(
2007
).
Genome-wide atlas of gene expression in the adult mouse brain
.
Nature
445
,
168
-
176
.
Li
,
W.
,
Wang
,
F.
,
Menut
,
L.
and
Gao
,
F.-B.
(
2004
).
BTB/POZ-zinc finger protein abrupt suppresses dendritic branching in a neuronal subtype-specific and dosage-dependent manner
.
Neuron
43
,
823
-
834
.
Li
,
Q.
,
Ha
,
T. S.
,
Okuwa
,
S.
,
Wang
,
Y.
,
Wang
,
Q.
,
Millard
,
S. S.
,
Smith
,
D. P.
and
Volkan
,
P. C.
(
2013
).
Combinatorial rules of precursor specification underlying olfactory neuron diversity
.
Curr. Biol.
23
,
2481
-
2490
.
Li
,
H.-H.
,
Kroll
,
J. R.
,
Lennox
,
S. M.
,
Ogundeyi
,
O.
,
Jeter
,
J.
,
Depasquale
,
G.
and
Truman
,
J. W.
(
2014
).
A GAL4 driver resource for developmental and behavioral studies on the larval CNS of Drosophila
.
Cell Rep.
8
,
897
-
908
.
Ludlow
,
C.
,
Choy
,
R.
and
Blochlinger
,
K.
(
1996
).
Functional analysis of Drosophila and mammalian cut proteins in flies
.
Dev. Biol.
178
,
149
-
159
.
Matthews
,
B. J.
,
Kim
,
M. E.
,
Flanagan
,
J. J.
,
Hattori
,
D.
,
Clemens
,
J. C.
,
Zipursky
,
S. L.
and
Grueber
,
W. B.
(
2007
).
Dendrite self-avoidance is controlled by dscam
.
Cell
129
,
593
-
604
.
Merritt
,
D. J.
and
Whitington
,
P. M.
(
1995
).
Central projections of sensory neurons in the Drosophila embryo correlate with sensory modality, soma position, and proneural gene function
.
J. Neurosci.
15
,
1755
-
1767
.
Moore
,
A. W.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2002
).
hamlet, a binary genetic switch between single- and multiple- dendrite neuron morphology
.
Science
297
,
1355
-
1358
.
Morcillo
,
P.
,
Rosen
,
C.
and
Dorsett
,
D.
(
1996
).
Genes regulating the remote wing margin enhancer in the Drosophila cut locus
.
Genetics
144
,
1143
-
1154
.
Morin
,
X.
,
Daneman
,
R.
,
Zavortink
,
M.
and
Chia
,
W.
(
2001
).
A protein trap strategy to detect GFP-tagged proteins expressed from their endogenous loci in Drosophila
.
Proc. Natl. Acad. Sci. USA
98
,
15050
-
15055
.
Neumann
,
C. J.
and
Cohen
,
S. M.
(
1998
).
Boundary formation in Drosophila wing: Notch activity attenuated by the POU protein Nubbin
.
Science
281
,
409
-
413
.
Paumard-Rigal
,
S.
,
Zider
,
A.
,
Vaudin
,
P.
and
Silber
,
J.
(
1998
).
Specific interactions between vestigial and scalloped are required to promote wing tissue proliferation in Drosophila melanogaster
.
Dev. Genes Evol.
208
,
440
-
446
.
Pfeiffer
,
B. D.
,
Jenett
,
A.
,
Hammonds
,
A. S.
,
Ngo
,
T.-T. B.
,
Misra
,
S.
,
Murphy
,
C.
,
Scully
,
A.
,
Carlson
,
J. W.
,
Wan
,
K. H.
,
Laverty
,
T. R.
, et al. 
(
2008
).
Tools for neuroanatomy and neurogenetics in Drosophila
.
Proc. Natl. Acad. Sci. USA
105
,
9715
-
9720
.
Philippidou
,
P.
and
Dasen
,
J. S.
(
2013
).
Hox genes: choreographers in neural development, architects of circuit organization
.
Neuron
80
,
12
-
34
.
Quinones-Coello
,
A. T.
,
Petrella
,
L. N.
,
Ayers
,
K.
,
Melillo
,
A.
,
Mazzalupo
,
S.
,
Hudson
,
A. M.
,
Wang
,
S.
,
Castiblanco
,
C.
,
Buszczak
,
M.
,
Hoskins
,
R. A.
, et al. 
(
2007
).
Exploring strategies for protein trapping in Drosophila
.
Genetics
175
,
1089
-
1104
.
Ryder
,
E.
,
Blows
,
F.
,
Ashburner
,
M.
,
Bautista-Llacer
,
R.
,
Coulson
,
D.
,
Drummond
,
J.
,
Webster
,
J.
,
Gubb
,
D.
,
Gunton
,
N.
,
Johnson
,
G.
, et al. 
(
2004
).
The DrosDel collection: a set of P-element insertions for generating custom chromosomal aberrations in Drosophila melanogaster
.
Genetics
167
,
797
-
813
.
Ryder
,
E.
,
Ashburner
,
M.
,
Bautista-Llacer
,
R.
,
Drummond
,
J.
,
Webster
,
J.
,
Johnson
,
G.
,
Morley
,
T.
,
Chan
,
Y. S.
,
Blows
,
F.
,
Coulson
,
D.
, et al. 
(
2007
).
The DrosDel deletion collection: a Drosophila genomewide chromosomal deficiency resource
.
Genetics
177
,
615
-
629
.
Satoh
,
D.
,
Sato
,
D.
,
Tsuyama
,
T.
,
Saito
,
M.
,
Ohkura
,
H.
,
Rolls
,
M. M.
,
Ishikawa
,
F.
and
Uemura
,
T.
(
2008
).
Spatial control of branching within dendritic arbors by dynein-dependent transport of Rab5-endosomes
.
Nat. Cell Biol.
10
,
1164
-
1171
.
Schindelin
,
J.
,
Arganda-Carreras
,
I.
,
Frise
,
E.
,
Kaynig
,
V.
,
Longair
,
M.
,
Pietzsch
,
T.
,
Preibisch
,
S.
,
Rueden
,
C.
,
Saalfeld
,
S.
,
Schmid
,
B.
, et al. 
(
2012
).
Fiji: an open-source platform for biological-image analysis
.
Nat. Methods
9
,
676
-
682
.
Schrader
,
S.
and
Merritt
,
D. J.
(
2000
).
Central projections of Drosophila sensory neurons in the transition from embryo to larva
.
J. Comp. Neurol.
425
,
34
-
44
.
Schrader
,
S.
and
Merritt
,
D. J.
(
2007
).
Dorsal longitudinal stretch receptor of Drosophila melanogaster larva – fine structure and maturation
.
Arthropod Struct. Dev.
36
,
157
-
169
.
Simmonds
,
A. J.
,
Liu
,
X.
,
Soanes
,
K. H.
,
Krause
,
H. M.
,
Irvine
,
K. D.
and
Bell
,
J. B.
(
1998
).
Molecular interactions between Vestigial and Scalloped promote wing formation in Drosophila
.
Genes Dev.
12
,
3815
-
3820
.
Smith
,
C. J.
,
Watson
,
J. D.
,
Spencer
,
W. C.
,
O'Brien
,
T.
,
Cha
,
B.
,
Albeg
,
A.
,
Treinin
,
M.
and
Miller
,
D. M.
(
2010
).
Time-lapse imaging and cell-specific expression profiling reveal dynamic branching and molecular determinants of a multi-dendritic nociceptor in C. elegans
.
Dev. Biol.
345
,
18
-
33
.
Smith
,
C. J.
,
O'Brien
,
T.
,
Chatzigeorgiou
,
M.
,
Spencer
,
W. C.
,
Feingold-Link
,
E.
,
Husson
,
S. J.
,
Hori
,
S.
,
Mitani
,
S.
,
Gottschalk
,
A.
,
Schafer
,
W. R.
, et al. 
(
2013
).
Sensory neuron fates are distinguished by a transcriptional switch that regulates dendrite branch stabilization
.
Neuron
79
,
266
-
280
.
Struhl
,
G.
and
Basler
,
K.
(
1993
).
Organizing activity of wingless protein in Drosophila
.
Cell
72
,
527
-
540
.
Sudarsan
,
V.
,
Anant
,
S.
,
Guptan
,
P.
,
VijayRaghavan
,
K.
and
Skaer
,
H.
(
2001
).
Myoblast diversification and ectodermal signaling in Drosophila
.
Dev. Cell
1
,
829
-
839
.
Sugimura
,
K.
,
Satoh
,
D.
,
Estes
,
P.
,
Crews
,
S.
and
Uemura
,
T.
(
2004
).
Development of morphological diversity of dendrites in Drosophila by the BTB-zinc finger protein abrupt
.
Neuron
43
,
809
-
822
.
Terriente
,
J.
,
Perea
,
D.
,
Suzanne
,
M.
and
Díaz-Benjumea
,
F. J.
(
2008
).
The Drosophila gene zfh2 is required to establish proximal-distal domains in the wing disc
.
Dev. Biol.
320
,
102
-
112
.
Tsubouchi
,
A.
,
Caldwell
,
J. C.
and
Tracey
,
W. D.
(
2012
).
Dendritic filopodia, Ripped Pocket, NOMPC, and NMDARs contribute to the sense of touch in Drosophila larvae
.
Curr. Biol.
22
,
2124
-
2134
.
Wu
,
S.
,
Liu
,
Y.
,
Zheng
,
Y.
,
Dong
,
J.
and
Pan
,
D.
(
2008
).
The TEAD/TEF family protein Scalloped mediates transcriptional output of the Hippo growth-regulatory pathway
.
Dev. Cell
14
,
388
-
398
.
Xiang
,
Y.
,
Yuan
,
Q.
,
Vogt
,
N.
,
Looger
,
L. L.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2010
).
Light-avoidance-mediating photoreceptors tile the Drosophila larval body wall
.
Nature
468
,
921
-
926
.
Yan
,
Z.
,
Zhang
,
W.
,
He
,
Y.
,
Gorczyca
,
D.
,
Xiang
,
Y.
,
Cheng
,
L. E.
,
Meltzer
,
S.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2013
).
Drosophila NOMPC is a mechanotransduction channel subunit for gentle-touch sensation
.
Nature
493
,
221
-
225
.
Ye
,
B.
,
Zhang
,
Y.
,
Song
,
W.
,
Younger
,
S. H.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2007
).
Growing dendrites and axons differ in their reliance on the secretory pathway
.
Cell
130
,
717
-
729
.
Ye
,
B.
,
Kim
,
J. H.
,
Yang
,
L.
,
McLachlan
,
I.
,
Younger
,
S.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2011
).
Differential regulation of dendritic and axonal development by the novel Kruppel-like factor Dar1
.
J. Neurosci.
31
,
3309
-
3319
.
Yeo
,
S. L.
,
Lloyd
,
A.
,
Kozak
,
K.
,
Dinh
,
A.
,
Dick
,
T.
,
Yang
,
X.
,
Sakonju
,
S.
and
Chia
,
W.
(
1995
).
On the functional overlap between two Drosophila POU homeo domain genes and the cell fate specification of a CNS neural precursor
.
Genes Dev.
9
,
1223
-
1236
.
Zecca
,
M.
and
Struhl
,
G.
(
2010
).
A feed-forward circuit linking wingless, fat-dachsous signaling, and the warts-hippo pathway to Drosophila wing growth
.
PLoS Biol.
8
,
e1000386
.
Zhang
,
L.
,
Ren
,
F.
,
Zhang
,
Q.
,
Chen
,
Y.
,
Wang
,
B.
and
Jiang
,
J.
(
2008
).
The TEAD/TEF family of transcription factor Scalloped mediates Hippo signaling in organ size control
.
Dev. Cell
14
,
377
-
387
.
Zheng
,
Y.
,
Wildonger
,
J.
,
Ye
,
B.
,
Zhang
,
Y.
,
Kita
,
A.
,
Younger
,
S. H.
,
Zimmerman
,
S.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
2008
).
Dynein is required for polarized dendritic transport and uniform microtubule orientation in axons
.
Nat. Cell Biol.
10
,
1172
-
1180
.
Zlatic
,
M.
,
Li
,
F.
,
Strigini
,
M.
,
Grueber
,
W.
and
Bate
,
M.
(
2009
).
Positional cues in the Drosophila nerve cord: semaphorins pattern the dorso-ventral axis
.
PLoS Biol.
7
,
e1000135
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information