Introduction
All behavior rests upon the ability of animals to detect variation in the internal and external environments. In multicellular animals, the detection of such variation is a function performed by sensory organs. With much of its external surface covered by many different classes of sensory organs, the fruit fly Drosophila melanogaster has long been used to investigate the mechanisms through which animals sense the world around them. Much attention has focused on the eyes, antennae, and maxillary palps, but the male Drosophila forelegs, which perform wide-ranging roles in locomotion, grooming, and courtship (e.g., [1–3]), display a distinct repertoire of sensory organs (Fig 1A and 1B). Internal mechanosensory receptors known as chordotonal organs sense proprioceptive stimuli around leg joints [4] and substrate-borne vibrations [5] (Fig 1A). Campaniform sensilla, singly innervated, shaftless sensors embedded in the cuticle, detect and relay cuticular strain, allowing for posture and intraleg coordination to be maintained [6–8] (Fig 1C). Mechanosensory bristles line the surface of the leg, detecting contact by deflection of an external, hair-like process [9,10] (Fig 1D). This organ class isn’t uniform: In the males of a subset of Drosophila species, including D. melanogaster, some mechanosensory bristles are heavily modified to generate a “sex comb,” an innovation critical for male mating success [11–14] (Fig 1B). Finally, the foreleg also contains chemosensory taste bristles, which are sexually dimorphic in number and innervated by both multiple gustatory receptor neurons (GRNs) and a single mechanosensory neuron [15–17] (Fig 1E). As in other parts of the body, such as the labellum (e.g., [18]), a degree of functional diversity exists between chemosensory taste bristles on the legs. The tunings and sensitivities of these bristles to a wide panel of tastants vary in relation to both the pair of legs on which they’re housed and their position within a given leg [19]. This variation is, at least in part, achieved by restricting the expression of certain gustatory receptors to subsets of taste bristles [3,19]. A level below the bristles themselves, the multiple GRNs that innervate each bristle appear to perform distinct functions. Three distinct GRN classes involved in the detection and evaluation of conspecifics have been resolved in the leg, each of which makes a critical contribution to normal sexual behavior [20–22]. Ultimately, unraveling how this varied sensory apparatus is constructed through development and identifying the molecular basis of specialization in each sensory organ remain central objectives of developmental neurobiology.
[Figure omitted. See PDF.]
Fig 1. The first tarsal segment of the male Drosophila melanogaster foreleg carries multiple functionally and structurally distinct sensory organs.
(A) Anatomy of the Drosophila melanogaster foreleg. The first tarsal segment (ta1), the focal region of this study, is distal to the tibia. Two chordotonal organs (COs) are present outside of the tarsal segments (approximate positions shown in purple). One is situated in the proximal femur (FeCO) and the other in the distal tibia (tCO) [4,5]. (B) The ta1 of the D. melanogaster foreleg is enriched for a range of functionally and structurally diverse sensory organs. This region has the highest concentration of mechanosensory bristles of any part of the leg. Here, mechanosensory bristles are arranged in transverse rows on the ventral side, an arrangement thought to aid in grooming, and longitudinal rows on the anterior, dorsal, and posterior sides [9]. In males, the most distal transverse bristle row is transformed into the sex comb: The mechanosensory bristles, now “teeth,” are modified to be thicker, longer, blunter, and more heavily melanized, while the whole row is rotated 90° [11,12]. Males also show a sex-specific increase in the number of chemosensory taste bristles in ta1, bearing approximately 11 compared to the female’s approximately 7 [16]. Three campaniform sensilla are present in ta1, two on the dorsal distal end of ta1 and one on the proximal ventral side [Ta1GF and Ta1SF, respectively, using the nomenclature of [8]; no campaniform sensilla are present in the distal tibia, ta2, or proximal ta3. (C-E) Campaniform sensilla, mechanosensory bristles, and chemosensory bristles are all composed of modified versions of four core cell types: a socket (or “tormogen”), shaft/dome (or “trichogen”), sheath (or “thecogen”), and neuron [186]. The shaft and socket construct the external apparatus that provides the point of contact for mechanical or chemical stimuli and form a subcuticular lymph cavity that provides the ion source for the receptor current [142,187]. The sheath has glia-like properties, ensheathing the neuron and, as is thought, providing it with protection [187]. Ultimately, however, the contributions of these nonneuronal cells to sensory processing remain poorly characterized [26]. (C) Campaniform sensilla detect strain in the cuticle. They are singly innervated and capped with a dome, rather than a hair-like projection, which extends across the surface of the socket cell [25]. The dendrite tip attaches to the dome cuticle [187]. (D) Mechanosensory bristles detect deflection of the hair-like projection. They are innervated by a single neuron, the dendritic projections of which terminate at the base of the shaft. Specific to this bristle class, the most proximal epithelial cell to the developing sense organ is induced to become a bract cell [12,80]. Bract cells secrete a thick, pigmented, hair-like, cuticular protrusion. (E) The chemosensory taste bristles of the leg differ in their morphology from mechanosensory bristles, appearing less heavily melanized and more curved. They also house a pore at the terminus of the shaft and lack bracts. Each is innervated by a single mechanosensory neuron and 4 gustatory receptor neurons (GRNs) [16]. Figure created using Biorender.com.
https://doi.org/10.1371/journal.pbio.3002148.g001
The function of sensory organs and their tuning to particular stimuli is not only a product of the neurons that innervate them. In each case, the sensory organ is a composite of multiple distinct cell types and dependent upon the involvement of glia to effectively relay detected signals to the brain. Different organ classes appear to share a common developmental blueprint, such that, despite variation in their form and function, mechanosensory bristles, chemosensory taste bristles, and campaniform sensilla each contain 4 homologous cell types [23]. In the bristle lineage, these are the neurons, which may vary in number between different sensory organ classes (such as between the polyinnervated chemosensory and monoinnervated mechanosensory bristles), along with 3 sensory support cells: the trichogen (shaft or, in campaniform sensilla, the dome), tormogen (socket), and thecogen (sheath). These sensory support cells bear features that clearly define the sensory capabilities of the organ. For example, the elongated shafts of mechanosensory bristles support the deflection-based mechanism through which stimuli are detected [10], the pore at the tip of chemosensory taste bristles enables the receipt of nonvolatile compounds [18], and the elliptical shape of many campaniform sensilla confers sensitivity to the direction of cuticular compression and strain ([24]; reviewed by [25]). But beyond these morphological features, our understanding of the wider, organ-specific contributions that support cells make to the specific sensory capabilities of each organ class remains poor [26]. Yet there is clear potential for their broader involvement in defining an organ type’s capabilities, given both their close physical associations with the neurons and, at least in taste bristles, their role in producing the lymph fluid that bathes the dendrites of GRNs and which is central to tastant detection [27,28].
High-throughput single-cell RNA sequencing (scRNA-seq) technologies allow for the transcriptional profiles of many thousands of cells to be recorded from a single tissue. The advent of these technologies has precipitated an explosion of interest in cell type–specific patterns of gene expression. Over the last few years, “atlases” describing the cellular diversity of tissues [29–32], embryos (e.g., [33–35]), and whole adult animals (e.g., [36–38]) have been published for a variety of species. Through such work, regulators of development have been identified (e.g., [39,40]), novel cell types described (e.g., [41,42]), and the effects of age (e.g., [43,44]) and infection (e.g., [45,46]) on the gene expression profiles of individual cell types characterized. But in arthropods, tissues associated with the cuticle have presented a challenge to single-cell approaches because the cuticle prevents isolation of single cells from peripheral tissues without significant damage [38,47]. Although single-nuclei RNA-seq methods have been used in such tissues (e.g., [38]), scRNA-seq approaches generally offer substantially greater read and gene detection with reduced “gene dropout” and lower expression variability between cells [48]. Consequently, approaches that help characterize the transcriptomes of cuticle-associated cells are of significant value. In principle, the developmental window between pupa–adult apolysis—the separation of the pupal cuticle from the epidermis—and the formation of the adult cuticle (approximately 12 to 48 h after puparium formation (APF)) [49] provides a rare opportunity during which such cells might be accessible.
Here, we use scRNA-seq to profile the sensory organs of the male D. melanogaster foreleg at 2 developmental time points that follow soon after the specification of sensory organ cells (24 h and 30 h APF) [11]. Using a fine-scale dissection technique, we specifically target the first tarsal segment (the “basitarsus”) to maximize the detection of rare sensory organ types, including the campaniform sensilla, chemosensory taste bristles, and sex comb teeth. We begin by examining the transcriptomic landscape of the tissues in which the sensory organs reside, constructing a spatial reference map of epithelial cells based on intersecting axes of positional marker expression, resolving joint-specific gene expression, and characterizing the distinct repertoires of expressed genes in tendon cells, hemocytes, and bract cells. We then focus on the nonneuronal component of the nervous system, describing the complement of glial cells present in the region, identifying and visualizing wrapping glia, surface glia, and a novel axon-associated cell population that is negative for the canonical glia marker repo and appears to contribute toward the construction of the neural lamella. We then resolve and validate a combinatorial transcription factor code unique to the neurons of each of mechanosensory bristles, campaniform sensilla, chordotonal organs, and the sex comb. We further identify and validate a transcription factor code unique to 4 transcriptomically distinct GRN classes, including known male- and female-pheromone sensing neurons, and recover this same code in a published adult leg dataset. With these annotations in place, we link a wide range of genes, including receptors and membrane channels, to specific neuron classes. Finally, we detail the transcriptomic differences that distinguish between sensory organ support cells, both within a single organ class (e.g., sheaths versus sockets) and between classes (e.g., chemosensory sheaths versus mechanosensory sheaths).
Results
Homologous clustering of 24 h and 30 h transcriptomes
We generated 2 scRNA-seq datasets from freshly dissected male first tarsal segments using 10× Chromium chemistry. One sample comprised males collected at 24 h APF and the other at 30 h APF. After filtering based on cell-level quality control metrics (see Materials and methods), we recovered 9,877 and 10,332 cells in our 24 h and 30 h datasets, respectively (S1A Fig). In the 24 h dataset, the median number of genes and transcripts detected per cell was 2,083 and 11,292, respectively (S1B and S1D Fig). The equivalent values for the 30 h dataset were 1,245 and 5,050 (S1C and S1E Fig). We began our analysis by constructing separate UMAP plots for each dataset. The clustering pattern of the 30 h dataset largely recapitulated that of the 24 h dataset, which is to say that each cluster in the 24 h dataset had a clear homolog in the 30 h dataset (and vice versa) based on marker gene expression (Fig 2A–2J). Given this concordance, we opted to integrate our 2 datasets and subcluster our data to facilitate closer analysis of rare cell subtypes (see Materials and methods). First, we subclustered epithelial cells and then separated them into joint and nonjoint datasets (Fig 2K). Epithelial cells represented the major cell type in both datasets (70.7% in 24 h: joints = 16.5%, nonjoints = 54.1%; 70.2% in 30 h: joints = 13.6%, nonjoints = 56.5%; Fig 2L). We then subclustered the nonepithelial cells (Fig 2M) and separated them into neurons (based on the expression of fne; Fig 2O), sensory support cells (based on the expression of pros, nompA, Su(H), and sv; Fig 2Q), and the remaining nonsensory cells (Fig 2P). The relative proportions of cells in each of these classes was similar between the 2 datasets (24 h: 19.5% neurons, 38.8% sensory support, 41.8% nonsensory; 30 h: 17.1% neurons, 37.0% sensory support, 45.9% nonsensory; Fig 2N). We include our final annotations in Fig 2 and work through the supporting evidence throughout this paper, with an emphasis on sensory and glial cell types.
[Figure omitted. See PDF.]
Fig 2. Clustering and iterative subsetting of integrated 24 h and 30 h APF scRNA-seq datasets identifies tarsal cell types with high resolution.
(A-J) UMAP plots showing cell clustering in the 24 h (A-E) and 30 h (F-J) datasets separately. Expression of a series of cluster markers is overlaid on the full 24 h (B-E) and 30 h (G-J) dataset UMAPs. At this resolution, each higher-level cluster in the 24 h dataset has a clear homolog in the 30 h dataset based on a selected subset of marker genes and vice versa. fne for neurons; ct for nonepithelial cells; NimC4 for hemocytes; aos for bracts; Sox100B and repo for different subtypes of glia and axon-associated cells; Su(H) for socket cells; sv for shafts and sheaths. (K) The central UMAP shows the clustering pattern observed in an integrated dataset containing just the epithelial joint and nonjoint cells from both 24 h (gold dots) and 30 h (green dots) samples. Joints are circled with a dashed light gray line, nonjoints with a dashed dark gray line. Reclustering the nonjoint and joint cells gave rise to the 2 flanking UMAP plots. See Fig 3 for details on how the annotations were determined. (L) The number of cells in the postfiltration, doublet-removed epithelial joint (yellow), epithelial nonjoint (gray), and nonepithelial cell (blue) datasets, plotted separately based on which sample (24 h APF or 30 h APF) the cells originated from. Numerical data with cell barcodes are listed in S1 Data. (M) UMAP showing the clustering pattern observed in an integrated dataset containing all nonepithelial cells from both the 24 h (gold dots) and 30 h (green dots) samples. Three major subsets of cells are grouped by colored shapes: neurons, nonsensory cells, and sensory support cells. (N) The number of cells in the postfiltration, doublet-removed sensory support (gold), neuron (navy), and nonsensory (pink) datasets, plotted separately based on which sample the cells originated from (24 h APF or 30 h APF). Numerical data with cell barcodes are listed in S2 Data. (O-Q) UMAPs showing the clustering pattern observed in integrated datasets containing all neurons (O), nonsensory cells (P), and sensory support cells (Q) from both the 24 h (gold dots) and 30 h (green dots) samples. See Figs 4–8 for details on how the annotations were determined. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g002
Transcriptomic divergence between the tibia/tarsus and intertarsal joints
Joint cells separated from the main body of epithelial cells in our initial epithelial clustering analysis (circled in Fig 3A and 3B). These cells showed enriched expression of genes with known involvement in the formation of or localization at joints, including drm, nub, and TfAP-2 (Fig 3A and 3B) [50–52]. After subclustering these cells (Fig 3C and 3D), we used the nonoverlapping expression of nub and TfAP-2 to identify subregions of the joint. We observed expression of GFP-tagged TfAP-2 [53] in both the tibia/tarsal and intertarsal joints, with somewhat weaker staining in the former (Fig 3E). In contrast, expression of nub-GAL4 was restricted to the joint-adjacent region of the distal tibia (Fig 3E) (see also [54,55]). One of our joint clusters was TfAP-2+/nub− and additionally positive for bab2, a gene with a documented tarsus-restricted expression profile (Fig 3A–3D) [56,57]. The other was divided into separate TfAP-2+ and nub+ domains. Based on our imaging, we propose that the TfAP-2+/nub− cluster corresponds to the ta1/ta2 joint, while the other corresponds to the tibia/ta1 joint. The tibia/ta1 joint can be further subdivided into the nub+ proximal and TfAP-2+ distal regions (Fig 3F; see S2A Fig for details of the high mt% cluster).
[Figure omitted. See PDF.]
Fig 3. Single-cell sequencing recovers positional information in the leg epithelium.
(A, B) UMAP plots of the integrated epithelial joint and nonjoint dataset overlaid with the expression of (A) drm (red) and bab2 (blue), and (B) nub (red) and TfAP-2 (blue). The 2 major joint clusters are circled and can be distinguished from one another based on the expression of these 4 genes. bab2 is known to be restricted to the tarsus starting with the distal first tarsal segment [56,57], providing one means through which to distinguish between the tibia and intertarsal joints. (C, D) As in (A) and (B) but with a UMAP plot of just the joint cells. (E) Confocal images showing 24 h pupal legs. On the left is a leg from a TfAP-2-GFP male. Staining is concentrated at the joints, which are each marked with a white triangle. Staining appears stronger at the intertarsal joints compared to the tibia/ta1 joint, consistent with the expression pattern of TfAP-2 in the scRNA-seq data. On the right is a leg from a nub-GAL4 > UAS-GFP.nls male. Staining is concentrated in the distal tibia, proximal to the tibia/ta1 joint (marked with a white arrow). Note that some nonspecific staining from contaminating fat body is present in ta1 and ta2. (F) Joint UMAP with clusters identified through shared nearest neighbor clustering and annotated based on the data presented in (A-E). For details of the high mt. % cluster, see S2A Fig. (G) Dot plot of a selection of top marker genes for each of the joint clusters given in F (excluding the high mt. % cluster). Marker genes were identified by comparing each cluster to the remaining joint clusters. Dot size reflects the number of cells in the cluster in which a transcript for the marker gene was detected, while color represents the expression level. (H, I) Expression of a selection of top marker genes identified in the analysis presented in G overlaid on the joint UMAP plot. (H) sob (red) and Ser (blue). (I) fj (red) and Lim1 (blue). (J-M) UMAP plots of the nonjoint epithelial cells overlaid with markers of spatial identity. Both (J) and (K) show expression of dorsal (red: bi and dpp) and ventral (blue: H15 and wg) markers. (L) shows anterior (blue: ci) and posterior (red: hh) markers. (M) shows proximal (blue: bab2) and distal (red: rn) markers. As is clear from the expression patterns, separation based on spatial markers is apparent for each axis, although stronger for dorsal–ventral and anterior–posterior than proximal–distal. This is likely due to us recovering only a small fraction of the proximal–distal axis by focusing in on just a single tarsal segment. (N) UMAP plot of the nonjoint epithelial cells colored by cluster identity as determined through shared nearest neighbor clustering. Spatial axes are illustrated by arrows based on the expression data presented in (J-M). (O) A dot plot showing the expression of positional markers across each cluster given in (N). Clusters are assigned to regions based on the positional gene expression signature they display. (P) A dot plot of the top markers for each cluster given in (N, O). Marker genes were identified by comparing each cluster to the remaining nonjoint epithelial clusters. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g003
To understand the broader transcriptomic differences between joint regions, we tested for differential gene expression in each cluster compared to all other clusters in our joint dataset (a selection is given in Fig 3G). We recovered several genes with known roles in joint formation, including the odd-skipped family transcription factors drm and sob (Figs 3A, 3H, S2B and S2C). These were enriched at the interface between the proximal and distal tibia/ta1 clusters, consistent with their known absence from the upper tarsal segments [52]. We also looked at 2 further members of the same gene family: odd, which showed a similar expression pattern but was present in fewer cells, and bowl, which was widely expressed among the joint clusters and in the wider body of epithelial cells (S2D and S2E–S2G Fig). We observed 2 major patterns among the top differentially expressed genes (DEGs) defining the proximal region of the tibia/ta1 joint. First, genes that appeared widespread among epithelial cells, but which were excluded from the other joint clusters (e.g., CG1648 and Ser; S2H and S2I Fig). Second, and more commonly, there were genes that showed strong specificity to the proximal tibia/ta1 joint (e.g., Lim1 –Fig 3I–nub, trh, caup, ara, pdm2, CG2016, and svp; S2J–S2P Fig). To the best of our knowledge, no role for pdm2 in leg development has been characterized, but it is believed to have evolved via duplication of nub, to which it is adjacent in the genome [58]. Our data suggest that the two are coexpressed, a conclusion that’s further supported by the recent discovery that the 2 genes share enhancers in the wing [59]. In contrast to the distinctiveness of the proximal tibia/ta1 expression profile, there was greater overlap in the DEGs for the distal tibia/ta1 and ta1/ta2 joints, with fewer cluster specific genes (Fig 3G; e.g., fj, Fig 3I; for UMAPs and discussion, see S2Q–S2V and S3A–S3R Figs). This suggests that while the transcriptomic distinctiveness of the proximal tibia/ta1 region is dominated by qualitative differences in expression, the more distal joint clusters show a more quantitative signal.
Epithelial cells express a signature of anatomical position
We next turned to the largest portion of our dataset, the nonjoint epithelial cells. Here, we observed clear separation between dorsal and ventral cells. This separation was clearest when mapping the expression of H15 (ventral) and bi (dorsal) (Fig 3J). wg (ventral) and dpp (dorsal) showed a similar, albeit weaker separation (Fig 3K). Unlike the joints, anterior–posterior separation was also clear, as delineated by the expression of ci (anterior) and hh (posterior) (Fig 3L). A weaker signature of proximal–distal separation could also be discerned from the expression of bab2, which is absent from the tibia and increases in expression between ta1 and ta2 (reviewed in [60]) (Fig 3M). We also detected localized expression of rn, the expression of which in ta1 is limited to the distal region [61] (Fig 3M). Recovery of spatial patterning in epithelial cells has recently been demonstrated in Drosophila wing imaginal disc scRNA-seq data [62,63]. But unlike in wing discs, we find that in this region of the leg the anterior–posterior signature is stronger than the proximal–distal, which may be due to our sequencing only a small fraction of the proximal–distal axis (i.e., just one tarsal segment). We then used the intersecting axes of positional marker expression as a spatial reference map to assign clusters to regions of the dissected leg tissue (Fig 3N and 3O). We tested for DEGs by comparing each region to the remainder (Fig 3P). Many DEGs showed signatures of localized up-regulation rather than cluster-specific expression, as would reasonably be expected from a tissue composed of a single cell type (exceptions include lbl, CG13064, CG13065, and CG13046; S3S–S3V Fig). But the cluster enriched for the distal ta1 marker rn exhibited a more specific gene expression profile, including showing enriched expression of the effector of sex determination dsx (reviewed in [64]), consistent with the localization of the sex comb to this region [65,66]. Genes enriched here represent candidate components of the sex-specific gene regulatory network that drives sex comb rotation (S3W–S3AE Fig).
Pupal leg hemocytes form a uniform population
We identified a single hemocyte cluster based on enriched expression of He, Hml, srp, and Nimrod-type receptor genes (Figs 4C, 4E and S4A–S4N) [67–69]. In our differential gene expression analysis, we observed strongly hemocyte-enriched expression of genes including CG31777, CG31337, CG3961, CG14629, CG4250, Mec2, CG42369, CG5958, Glt, and CG10621 (Fig 4B; see S4X–S4AG Fig for UMAPs of the first 5 genes). To test for the presence of hemocyte subtypes, we ran a wide panel of recently identified lamellocyte, crystal cell, and plasmatocyte subtype-specific genes against our data [70]. However, we saw no obvious subclustering in relation to these genes: They were either widely expressed among hemocyte cells, too patchily expressed to reflect a clear subpopulation, or absent from our dataset (S4O–S4W Fig). The absence of clear subclustering may reflect the rarity of these hemocyte subpopulations, that recovered cells are insufficiently differentiated at these time points to discriminate subclasses, that these notably fragile cells [71] lyse during tissue dissociation, or point to differences between the larval and pupal immune cell repertoire, for which there is some evidence [72,73].
[Figure omitted. See PDF.]
Fig 4. The tarsus contains several types of nonsensory, nonepithelial cells.
(A) Annotated UMAP plot of nonsensory cells. Cell labels provided in purple indicate populations that are discussed in this figure. Those in black are discussed in Fig 5. (B) Dot plot of the expression of top differentially expressed genes identified through comparisons between each named cluster and all remaining clusters in (A). (C-F) The nonsensory UMAP shown in (A) overlaid with expression of key marker genes for each cluster. Note that dsx (D) and dally (F) are expressed in a distinct subset of bract cells, which likely corresponds to sex comb bracts. (G) 24 h APF male pupal upper tarsal segments showing staining from 1151-GAL4 > UAS-mCherry.nls (magenta) and the neuronal marker anti-Futsch (green). 1151-GAL4 marks tendons [84]. The arrangement of tendon cells is clearly distinct from the paired nerve fibers that run along the same axis. (H-J) 24 h APF first tarsal segment and distal tibia from an 1151-GAL4 > UAS-mCherry.nls (magenta) male counterstained with anti-Vvl (green). Costaining is clearer in the levator and depressor tendons at the distal tibia/ta1 joint (marked with an arrow) than in the long tendon, which extends along the proximal–distal axis of the tarsal segments. This may be due to the greater concentration of tendon cells in this region and difficulties distinguishing between anti-Vvl staining in mechanosensory bristle cells (see Fig 6Q–6S) and tendon cells. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g004
The induction of bract identity is accompanied by a transcriptomic shift away from epithelial cell profiles
Most, but not all, mechanosensory bristles on the legs are associated with a bract cell [9,49]. We identified bracts based on the expression of the transcription factor pnt and aos, an EGF inhibitor selectively expressed in cells assuming bract fate [74–76]. Among our nonsensory clusters, we find 2 enriched for aos and pnt (Figs 4D, 4F, and S5A–S5D). Differential gene expression analysis comparing among our nonsensory cell clusters showed that the top markers of the major bract cluster also showed elevated expression in the minority cluster but that the top markers of the minority cluster were more specific in their expression (Fig 4B). The top DEGs for the minority cluster included several genes that were among the top markers of the putative sex comb bearing region identified in our epithelial cell analysis (rn, dsx, and dally; Fig 4D and 4F). Thus, these cells likely correspond to sex comb bracts. Given that bract identity is induced in epithelial cells, rather than emerging through the sensory organ precursor lineage, the natural comparison to make to identify putative determinants of bract fate is to compare bract cells with other epithelial cells [12,74,77–80]. Comparing the 2 bract clusters with the nonjoint epithelial cells, we observed several genes that were highly enriched in bracts and largely absent from epithelial cells (S5E Fig). It was common to find expression of some of these genes (e.g., CG33110, Nep2, CG32365, neur; S5F–S5P Fig) in bristle shaft and socket cells, which, considering the short bristle hair-like protrusion that bracts develop, may reflect their partially overlapping morphological characteristics. Ultimately, the distinct expression profile of the bracts suggests that induction of this identity in an epithelial cell is followed by remodeling of its transcriptome.
Tendon cells express the POU transcription factor vvl, but not sr, between 24 h and 30 h APF
We identified a cluster of tendon cells based on the expression of Tsp, tx, and the joint marker drm (Fig 4B and 4E) [81–83]. To visualize the anatomical distribution of tendon cells in the focal leg region, we crossed the verified tendon marker line 1151-GAL4 [84] to UAS-mCherry.nls and counterstained with an antibody against the neuronal marker Futsch to distinguish between tendons and axonal trunks (Fig 4G). The dissected region contains the “long tendon,” which runs along the proximal–distal axis of the tarsal segments, as well as the distal portion of the “tarsus levator” and “tarsus depressor” tendons, which are housed in the distal tibia [84]. One of the top DEGs for our Tsp+/tx+ cells was the POU homeobox transcription factor vvl (Fig 4C). When counterstaining 1151-GAL4>UAS-mCherry.nls legs with an antibody raised against Vvl, we observed clear costaining, supporting a tendon cell identity for this cluster (Fig 4H–4J). Of the top DEGs we identified for tendon cells, none were entirely specific to this cluster when looking across all cells in the dataset. A common pattern was to see localized expression among epithelial cells (e.g., drm, Tsp, CG13003) or among sockets and shaft cells (e.g., tx and CG42326) (S6A–S6R Fig). Beyond the top 10 tendon cell DEGs, we detected significantly enriched expression of trol, which encodes the extracellular matrix proteoglycan Perlecan (validated using trol-GAL4; S6S–S6V Fig). Surprisingly, we did not detect enriched expression of the tendon-specifying transcription factor sr in this cluster, nor its tendon-specific downstream targets slow and Lrt [85,86] (S6W–S6AE Fig). Their absence may reflect the developmental time points that we sequenced or differences between this tibia/tarsus tendon population and more commonly studied populations in the embryo [87] and wings [88].
Glial cells in the developing first tarsal segment have noncanonical expression profiles
Three major classes of glia have been described in the leg: perineural and subperineural glia (which collectively comprise the surface glia), and the PNS-specific wrapping glia [89]. To identify these populations in our dataset, we mapped the expression of the canonical glia marker repo, which is thought to be expressed in all lateral/embryonic glial cells except for a subset of specialized wrapping glia in the CNS known as midline glia [90–94]. We observed 2 repo+ clusters, one of which was svp+ and the other Oaz+ (Fig 5A–5E). Previous work has shown that Oaz specifically labels wrapping glia in larval peripheral nerves, where it is coexpressed with Repo [95]. In our stainings, we observed that anti-Oaz labeled a small number of repo-GAL4+ cells (Fig 5N–5P). Beyond Oaz, however, the expression patterns of the repo+ cells were atypical with respect to known marker genes. The subperineural glia marker genes moody and Gli were widely expressed across repo+ cells, including in the Oaz+ putative wrapping glia cells (Fig 5H and 5I) [89,96]. Conversely, the wrapping glia markers nrv2 and Ntan1 weren’t restricted to the Oaz+ cluster (Fig 5J and 5K), while the perineural glia marker Jupiter was widely detected across all nonsensory cells (S8F Fig). Indeed, genes previously shown to be enriched in surface glia [97] were similarly represented in the DEGs of each cluster (svp+ glia: 20/285, approximately 7.0%; Oaz+ glia: 14/223, approximately 6.3%; top DEGs are given in Fig 5B, with an expanded list in S7A Fig).
[Figure omitted. See PDF.]
Fig 5. Noncanonical expression patterns in leg glia and a new cell type associated with the neural lamella.
(A) Annotated UMAP plot of nonsensory cells. While the Oaz+ and svp+ glia cells express the canonical glia marker repo, the Sox100B+ cells do not. Cell labels provided in purple indicate populations that are discussed in this figure. Those in black are discussed in Fig 4. (B) Dot plot of the expression of repo and gcm, the canonical glia markers, along with top differentially expressed genes identified through comparisons between each of Sox100B+ glia, Oaz+/repo+ glia, and svp+/repo+ glia against all the clusters named in (A). Of these genes, sr is known to induce tendon cell fate, and to the best of our knowledge, no functions have previously been reported for sr in glia. Unc-5 and Fas2 are both required for glial migration (reviewed in [93]). Genes identified as differentially expressed through more targeted between-glia comparisons are given in S7A Fig. (C-G) UMAP plots of the subsetted Sox100B+ cells, Oaz+/repo+ glia, and svp+/repo+ glia from (A) overlaid with the expression of a series of top marker genes for each cluster. (H-M) The UMAP plot shown in (A) overlaid with the expression of a series of glia markers. moody and Gli are subperineural glia markers, nrv2 and Ntan1 are wrapping glia markers, apt is a surface glia marker (i.e., a marker of both perineural and subperineural glia), and gcm is the upstream determinant of glial identity [89,96]. (N-P) 24 h APF legs from repo-GAL4>UAS-mCherry.nls males counterstained with anti-Oaz, a marker of wrapping glia [95]. Oaz+ cells are denoted by an arrow in the left-hand image. In Oaz+ cells at 24 h APF, the repo-GAL4+ signal was often weak and in one of the 4 legs we imaged, the one shown here, we observed a single Oaz+ cell that appeared repo-GAL4−. This is the topmost of the Oaz+ cells to which an arrow is pointing. (Q, R) Brain, ventral nerve cord, and leg discs (Q) and a close-up of a leg disc (R) from repo-GAL4>UAS-mCherry.nls males counterstained with anti-Sox100B. Note how repo-GAL4+ cells can be seen migrating into the disc from the CNS, while Sox100B+ cells appear to originate within the disc itself. (S-U) 24 h APF male upper tarsal segments from repo-GAL4 > UAS-mCherry.nls counterstained with anti-Sox100B. Both show a similar, but nonoverlapping, distribution of stained cells. (V-X) 24 h APF male upper tarsal segments from Lim1-GAL4 > UAS-mCD8::GFP counterstained with anti-Futsch. Above the tibia/ta1 joint, Lim1-GAL4 was expressed in the epithelial cells of the distal tibia, as predicted by our epithelial joint analysis (Fig 3I). Below the joint, the staining surrounded and spanned the distance between the 2 central axon trunks into which the sensory neuron axons project. (Y-AA) 24 h APF male upper tarsal segments from repo-GAL4 > UAS-mCD8::GFP counterstained with anti-Futsch. Unlike the Lim1-GAL4, repo-GAL4 staining does not span the gap between the 2 axon trunks with which it is closely associated, and cell bodies are clearly seen branching away from the fibers. (AB-AG) The UMAP plot shown in (A) overlaid with the expression of a series of top markers identified in this study. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g005
Despite these surprisingly broad expression patterns of established marker genes, 2 features point to known identities. First, svp-lacZ is known to be expressed in all surface glia [91]; among nonsensory cells in our data, svp was restricted to the Oaz−/repo+cells (Fig 5D). Second, among repo+ cells, the surface glia markers apt and Gs2 [89] appeared similarly restricted to the Oaz− cells (Figs 5L and S8G). Collectively, this suggests that our Oaz+ population correspond to wrapping glia, while the svp+ population corresponds to surface glia. This latter population appears heterogeneous, with localized up-regulation of Gli detectable (Fig 5I). Several processes may underlie this heterogeneity. One possibility is that it reflects developmental staging differences among surface glia. This is supported by the localized expression of the upstream determinant of glial identity, gcm, which is known to act early in development (Fig 5M) [98]. But another possibility is that this cluster includes a mix of both perineural and subperineural glia. Perineural glia are far more numerous than subperineural glia in the leg [89], so any we recover may present as a subregion within a surface glia cluster otherwise dominated by perineural glia. Finally, surface glia may naturally be heterogenous in their expression profiles, as has been suggested by others [89].
A novel cell type associated with the neural lamella
Beyond the repo+ clusters, we observed a repo− population that was enriched for expression of the transcription factors Sox100B and Lim1 (Fig 5F and 5G) and expressed the midline glia marker wrapper [99–101] (Fig 5AB). To verify the mutually exclusive expression patterns of repo and Sox100B, we counterstained repo-GAL4 > UAS-mCherry.nls legs with an anti-Sox100B antibody across multiple time points. First, we looked at 0 h APF leg discs (Fig 5Q and 5R). Here, we observed no overlap in expression but noted a difference in the behavior of these 2 cell populations: While repo-GAL4+ cells could be seen migrating into the leg disc, as CNS-derived glia are known to do [89], we detected no such migratory behavior in the Sox100B+ cells. To determine whether Sox100B+ cells migrate into the disc at an earlier time point or originate within the disc itself, we next looked at L2 and L3 larval discs (S7B and S7C Fig). We observed migration of repo-GAL4+ cells across both time points. In contrast, Sox100B+ cells were present in the discs only in L3 larvae and showed no clear indication of migration. We therefore conclude that Sox100B+ cells originate within the leg disc itself.
At 24 h APF, we again detected no overlap in repo-GAL4 and anti-Sox100B staining in the legs. Despite being repo-GAL4−, the nuclei of Sox100B+ cells occupied glia-like positions in relation to the axon trunks at this time point (Fig 5S–5U). A glia-like association between Sox100B+ cells and the neuropil was also evident in the larval (S7B Fig) and pupal (S7D Fig) ventral nerve cord. In 24 h APF legs, the morphology and neuronal associations of Sox100B+ and repo-GAL4+ cells were clearly distinct. Expressing a membrane-tethered form of GFP under the control of a GAL4 driver for one of the top markers of the Sox100B+ cluster (Lim1-GAL4; [102]), we observed that these cells appear to surround the axon trunks (Fig 5V–5X). When compared to the equivalent staining for repo-GAL4, the staining around the axon trunks in Lim1-GAL4 > UAS-mCD8::GFP legs appeared larger in diameter, suggesting that it comprises a layer that is outer to that of the repo-GAL4+ cells (Figs 5Y–5AA, S8A, and S8B). To exclude the possibility that these cells correspond to myoblasts, we compared the distribution of Sox100B+ cells to those stained by an antibody raised against the myoblast marker Mef2 (S8C Fig) [103]. Unlike Sox100B+ cells, myoblasts were restricted to the tibia and absent from tarsal segments, arguing against a myoblast identity for Sox100B+ cells.
We next visualized a protein trap of one of the Sox100B+ cluster’s most specifically enriched markers, vkg, which encodes a subunit of the extracellular matrix component Collagen IV and which was essentially absent from the repo+ glia (Figs 5AC and S8D). The vkg::GFP staining pattern resembled Lim1-GAL4>UAS-mCD8::GFP: a broad ensheathing of the axon trunks that extended more laterally than the equivalent staining observed for repo-GAL4 (S8D Fig). This provides further support for the “outerness” of these cells. vkg::GFP is known to label the neural lamella, a dense network of extracellular matrix that surrounds the central and peripheral nervous systems and which is required to help control their shape [96,104,105]. These features suggest that the Sox100B+ cells we identify here may be involved in the construction of the neural lamella. This role is thought to be performed by migrating hemocytes during embryogenesis [96,106] and consistent with this we find that leg hemocytes also express vkg, albeit less strongly (Fig 5AC). But beyond this, the only other clear similarity we detect between our hemocyte and Sox100B+ cells was the expression of NimC3, which was highly expressed in hemocytes and showed low-level expression in Sox100B+ cells (S4K Fig). It’s therefore possible that migrating hemocytes work alongside Sox100B+ cells to construct the neural lamella. Consistent with this, we see both cluster-specific and overlapping expression of many extracellular matrix component genes: SPARC is expressed in Sox100B+ cells, hemocytes, and repo+ glia; trol shows low-level expression in Sox100B+ cells, hemocytes, and svp+/repo+ glia; Col4a1 and vkg are present in both Sox100B+ cells and hemocytes; and Pxn is hemocyte-specific (Figs 5AC and S8H–S8K). However, the position of Sox100B+ cells at the outer layer of glia, along with their strong enrichment of the canonical neural lamella marker vkg, suggest that this cell type may play the primary role in neural lamella synthesis during pupal development, with any contribution by the hemocytes being secondary.
repo+ glia and Sox100B+ cells express distinct cell–cell communication gene repertoires
Comparing between the repo+ glia and Sox100B+ cells, we observed cluster-specific expression of beaten path family genes. beat-IIa and beat-IIb were restricted to the Sox100B+ cluster, while beat-IIIc was enriched in repo+ cells (Fig 5AD and 5AF). beaten path genes are thought to act as neuronal receptors for sidestep gene family ligands expressed in peripheral tissues [107]. Thus, the differential expression of different subsets of beaten path family genes between repo+ and Sox100B+ cells point both to the importance of these genes in the nonneuronal component of the nervous system and to cell type–specific patterns of between-cell communication in the developing nervous system. These beat genes have been recorded elsewhere in the fly: In the visual system, beat-IIb is expressed in L3 and L4 lamina neurons, the glia beneath L5, and in the lamina neuropil, while beat-IIIc is expressed in a subset of retinal neurons [108]. Other cell communication pathway elements also showed cell type specificity. For example, we observed that the fibroblast growth factor (FGF) ligand ths was, among glia, largely restricted to svp+/repo+ cells (Fig 5AG). FGF signaling is known to underlie aspects of neuron–glia communication in Drosophila, although in these cases, the source of Ths is neuronal. In one example, Ths is thought to act in neurons as a directional chemoattractant for the migration of astrocytes and the outgrowth of their processes [109]. In 2 others, the release of Ths from olfactory neurons directs ensheathing glia to wrap each glomerulus [110], while Ths in photoreceptor neurons induces differentiation of glia in the developing eye [111]. The expression of ths in one of our glia populations, specifically the population we believe to correspond to the surface glia, is therefore surprising. It’s possible that FGF-mediated interactions between different glia populations guide their concerted differentiation and the development of the close physical associations they form. Similar interactions with neurons may also help guide the growth of neuronal projections in the vicinity of these glia.
A combinatorial transcription factor code for sensory neurons
We recovered multiple distinct sensory neuron populations in our clustering analysis, each defined by the expression of a unique combination of transcription factors (Fig 6A and 6B). We recovered a similar clustering pattern in our analysis of male neurons from the Fly Cell Atlas (FCA) adult leg data (Fig 6G and 6H) [38]. In contrast to our pupal data, however, the FCA dataset is derived from all segments of all 3 pairs of legs and is single-nuclei, rather than single-cell. We failed to recover clear sex comb or MSNCB (mechanosensory neuron in chemosensory bristle) populations in the FCA data. But in their place, we recovered 3 populations apparently absent from our pupal dataset. These novel clusters were enriched for CG9650, a transcription factor we found to be enriched in joint and tendon cells, and showed cluster-specific, combinatorial expression of transcription factors, including erm and bab1 (Fig 6I and 6J). Consistent with signs of a joint identity, we believe these are likely to correspond to chordotonal neuron populations, a class that is absent from the first tarsal segment. The pupal and FCA neuron datasets did not integrate as well as our pupal datasets did to each other (S9I and S9J Fig). In light of the significant differences in sample preparation, age, cells versus nuclei, and dissected region between the datasets we opted to analyze them separately.
[Figure omitted. See PDF.]
Fig 6. Identification of a combinatorial transcription factor code for leg sensory neurons.
(A) Annotated UMAP plot of neuronal cells from the integrated 24 h AFP and 30 h APF first tarsal segment dataset. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. See S9A–S9H Fig for details on the TkR86C+ mechanosensory neurons. (B) Dot plot showing the expression of a series of canonical neuronal markers (elav, nSyb, and para) and transcription factors across the major neuron class clusters labeled in the UMAP given in (A). Each cluster expresses a unique combination. The dotted lines separate the canonical neuronal markers and then the chemoreceptor from mechanoreceptor organs transcription factor markers. (C-F) UMAP plot of neuronal cells from the integrated 24 h AFP and 30 h APF first tarsal segment dataset overlaid with the expression of members of the transcription factor code depicted in (B). (C) Note how the MSNCB cluster branching off from the top of the mechanosensory neuron population is negative for both vvl and pros. (D) Ets65A is present in all non-GRN populations in the UMAP, while an effector of sex differentiation, dsx, is expressed in GRNs, sex comb neurons, and MSNCBs. (E) fru, the other effector of sex differentiation, is enriched in 2 GRN populations and sex comb neurons, while eyg is restricted to campaniform sensilla neurons. (F) CG42566 is the only nontranscription factor plotted. It is a top marker of MSNCBs and its expression in both MSNCBs and GRNs contributed to this cluster’s chemosensory bristle annotation. ham is enriched in mechanosensory neuron classes and 2 GRN populations. (G) Annotated UMAP plot of male neuronal cells subsetted from the Fly Cell Atlas single-nuclei RNA-seq leg dataset [38]. Note the presence of 3 clusters, annotated as “putative chordotonal,” which are absent from the pupal dataset—chordotonal organs are not present in the upper tarsal segments. No clear MSNCB or sex comb clusters could be resolved in this dataset. (H) As (B) but for the male neuronal cells subsetted from the Fly Cell Atlas single-nuclei RNA-seq leg dataset. Only those clusters present in the pupal single-cell data are shown. (I) UMAP plot of male neuronal cells subsetted from the Fly Cell Atlas single-nuclei RNA-seq leg dataset overlaid with expression of the mechanosensory neuron marker vvl (blue) and a top marker of the putative chordotonal organs, the predicted transcription factor CG9650 (red). (J) A subset of (I), showing only the putative chordotonal clusters overlaid with expression of 2 transcription factors, bab1 (blue) and erm (red). (K-V) Confocal images of 24 h APF male first tarsal segments. (K-M) Mechanosensory bristles from elav-GAL4 > UAS-mCherry.nls (magenta) stained with anti-Pros (green). Two elav-GAL4+ cells are present per mechanosensory bristle, one of which, the sheath, is Pros+. elav-GAL4 expression in the sheath is likely due to the legs being imaged soon after the division of the common pIIIb progenitor cell from which they derive (see also [113] and S9K–S9M Fig). MSN, mechanosensory neuron. (N-P) Two chemosensory bristles (circled) from elav-GAL4 > UAS-mCherry.nls (magenta) stained with anti-Pros (green). Note that each bristle includes 4 Pros+/elav-GAL4+ cells (the gustatory receptor neurons), 1 Pros+/elav-GAL4− cell (the chemosensory sheath cell), and 1 Pros−/elav-GAL4+ cell (the MSNCB, mechanosensory neuron in chemosensory bristle). (Q-S) Two chemosensory (CS) bristles and 1 mechanosensory (MS) bristle from elav-GAL4 > UAS-mCherry.nls (magenta) stained with anti-Vvl (green). Note that anti-Vvl staining is entirely absent from the CS bristle including, therefore, the mechanosensory neuron (MSNCB) that innervates it. Conversely, anti-Vvl staining is observed in all 4 constituent cells of a MS bristle. (T-V) The same stainings performed in (Q-S) but centered on the sex comb. Anti-Vvl staining is present in both the neuronal (elav-GAL4+) and nonneuronal cells of the sex comb. The “central bristle,” which develops from the same bristle row as the sex comb is labeled. (W-Y) Confocal images of 48 h APF male first tarsal segments showing the expression of fru-GAL4 (magenta) and anti-Futsch (green). fru-GAL4 expression is restricted to the sex comb and chemosensory (CS) neurons. The later 48 h time point was used as fru-GAL4 was undetectable up until 40 h and weak up until 48 h. (Z) Confocal image of the first tarsal segment from a 24 h male from eyg-GAL4 > UAS-GFP.S65T. Campaniform sensilla are marked with asterisks. The axonal projections can be seen as parallel lines running either side of the central autofluorescence. Note that some nonspecific fat body staining is also present in this image. (AA) Confocal image of a distal first tarsal segment campaniform sensillum from a 24 h male where eyg-GAL4 is driving the expression of UAS-mCherry.nls. The top and bottom image in this panel show the same sensillum but with different levels of saturation to variously highlight the domed structure (top) and the individual cells of the organ (bottom). (AB) As (Z) but showing an adult haltere. Note that the staining is restricted to the campaniform sensilla field on the pedicel (“Ped.”) and apparently absent from the field on the scabellum (“Sca.”). Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g006
Unique combinations of the transcription factors vvl, pros, and fru distinguish between mechanosensory, chemosensory, and sex comb neurons
In both our pupal and the FCA data, the nonoverlapping expression of vvl and pros marked the highest order difference between neuron clusters (Fig 6B, 6C, and 6H). Among sensory organ cells, pros is known to be expressed in sheath cells [112] and has been detected in DA1, DL3, and VA1d olfactory receptor neurons in scRNA-seq data from antennae [47]. Consistent with sheath cell expression, we observed anti-Pros staining in 1 cell per mechanosensory bristle (Fig 6K). Surprisingly, however, in many cases the Pros+ cell was also positive for the canonical neuron marker elav (Fig 6L and 6M). We generally observed elav-GAL4 expression in 2 cells per bristle, despite mechanosensory bristles being singly innervated (Fig 6L and 6M). The expression of elav-GAL4 in the mechanosensory sheath cell likely reflects the early time point (24 h APF) at which we imaged. This conclusion was also reached by Simon and colleagues [113] after detecting elav expression in sheaths in 28 h APF mechanosensory bristles on the notum. Consistent with this conclusion, we observed a heterogenous mix of Pros+/elav-GAL4+ and Pros+/elav-GAL4− mechanosensory bristle sheaths in some legs, suggesting between-mechanosensory bristle variation in developmental stage (S9K–S9M Fig). Expression of elav beyond the neuron has now been noted several times: Its expression appears surprisingly broad and its neural specificity dependent on posttranscriptional repression outside of neurons [35,114].
Unlike in mechanosensory bristles, we detected multiple Pros+ cells per chemosensory bristle at 24 h APF (Fig 6N–6P). One of these was elav-GAL4−, consistent both with a sheath identity and, when contrasted against the heterogenous elav-GAL4 expression we observed among mechanosensory sheaths, the developmental timing differences that are known to exist between mechanosensory and chemosensory bristles: Chemosensory bristles are specified earlier than all but the largest mechanosensory bristles [115,116]. However, in contrast to mechanosensory bristles, anti-Pros staining in chemosensory bristles was not restricted to the sheath, extending to a further 4 elav-GAL4+ cells (Fig 6N–6P). Mirroring this arrangement, we detected 4 Pros+ populations in both the pupal and FCA data, suggesting that these correspond to 4 distinct GRN subtypes. Not all elav-GAL4+ cells in the chemosensory bristle were Pros+ though: We observed a single Pros−/elav-GAL4+ cell per bristle, which likely corresponds to the MSNCB (see below). Surprisingly, we failed to detect Poxn, which is known to be both necessary and sufficient for a chemosensory rather than mechanosensory fate, in GRNs in either dataset, which suggests that Poxn transcription may have ceased by 24 h APF [117–121].
In mechanosensory bristles, anti-Vvl marked all 4 of the constituent cell types (socket, shaft, neuron, and sheath) (Fig 6Q–6S). In contrast, no cells in chemosensory bristles were marked by anti-Vvl. Consequently, the mechanosensory neurons innervating mechanosensory and chemosensory bristles can be distinguished based on the presence of Vvl in the former and not the latter. In our pupal scRNA-seq data, we observe a cluster of vvl− cells branching off from the major vvl+ neuron population (Fig 6C). This cluster is otherwise positive for transcription factors present in the major mechanosensory population, such as Ets65A and ham (Fig 6D and 6F) and therefore likely corresponds to MSNCBs. Consistent with this, one of the top markers for the cluster, CG42566, was absent from the major mechanosensory population but expressed in 3 of the 4 GRN populations (Fig 6F). CG42566 remains largely restricted to GRNs in the adult FCA data (S9N Fig). Similar to MSNCBs, we observed an additional cluster branching off from the major mechanosensory population in the pupal data. However, this population was vvl+ and, uniquely among nonchemosensory neurons in the region, heavily enriched for fru. Our stainings suggest that these cells correspond to the sex comb neurons, which we observed to be both Vvl+ and fru-GAL4+ (Fig 6T–6Y; see also [122] for a previous report of fru expression in sex comb neurons). Collectively, these 3 transcription factors—pros, fru, and vvl—represent candidate high-level regulators of networks of downstream genes specific to the neurons of 3 major sensory organ classes: mechanosensory bristles, chemosensory bristles, and the sex comb.
Campaniform sensilla express the Pax family transcription factors eyg and toe
In our pupal data, we resolved a small cluster of cells enriched for the Pax family transcription factors eyg and toe. Expressing UAS-GFP under the control of eyg-GAL4, we observed staining in the regions of the first tarsal segment that correspond to the positions of the campaniform sensilla: 1 proximal organ and 2 distal organs [8] (Fig 6Z). Repeating the experiment with a nuclear-localizing UAS-mCherry, we detected expression in 4 cells within each tarsal campaniform sensillum, which presumably correspond to the neuron, sheath, socket, and dome cell (Fig 6AA). Although the expression of eyg and toe were relatively low in the adult nuclei campaniform sensilla neuron cluster, we observed eyg-GAL4 activity in both adult legs (S9O Fig) and in the adult haltere (Fig 6AA and 6AB). In the haltere, eyg-GAL4 activity was detectable in the field of campaniform sensilla on the pedicel, but not the scabellum, raising the possibility that there exist distinct subtypes of campaniform sensilla that express unique gene repertoires.
The legs contain 4 gustatory receptor neuron (GRN) classes, each expressing a unique combination of the transcription factors acj6, fru, nvy, and fkh
Our stainings showed that among neurons Pros was restricted to chemosensory bristles, suggesting that the multiple pros+ neuron clusters we detect in our scRNA-seq data represent subclasses of GRNs. Each GRN cluster expressed a unique combination of 5 transcription factors: pros+/acj6+/nvy+, pros+/acj6+/fkh+, pros+/acj6+/fru+, and pros+/fru+ (Fig 7A–7F). We validated these combinations using fru-GAL4 in conjunction with antibodies raised against Pros, Acj6, Nvy, and Fkh (Fig 7G–7AA). Multiple bristles are often closely associated within a single region of the leg, while the neurons themselves frequently overlap within a single bristle. Consequently, it wasn’t possible to definitively determine whether 1 cell of each GRN class was present in every ta1 bristle, but from our observations, this seems likely to be the case. Consistent with this, the numbers of cells recovered in each cluster generally appeared similar (Fig 7A). We recovered the same 4 populations, marked by the same transcription factor code, in the FCA full leg dataset, again recovering a similar number of cells in each GRN population (Fig 7AC). The expression of nvy was, however, far less extensive than in the pupal data, suggesting that expression drops off during later pupal development or that nvy is restricted to a subset of cells in this GRN class. Correspondence between the nvy+ cluster in the pupal and adult data was supported by additional marker genes, such as foxo and Fer1 (see below). Among neurons, the 4 GRN populations showed specific or enriched expression of Ir25a, Ir40a, Gluclalpha, RhoGAP102A, Snmp2, CG42540, CG13578, Tsp47F, and CG34342 (S10A–S10T Fig). Taken together, the recovery of the same 4 GRN classes across different time points, technologies, and dissected regions suggests that despite substantial between-bristle variation in receptor expression and sensitivity to given stimuli [19], just 4 core GRN classes might be present in the leg and that these classes are defined by combinatorial expression of a small set of transcription factors (Fig 7AB).
[Figure omitted. See PDF.]
Fig 7. Four gustatory receptor neuron (GRN) classes express a combinatorial transcription factor code and unique gene repertoires.
(A) Annotated UMAP of the pupal integrated neuron data. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. The number of cells in each GRN cluster is presented. The numbers are generally similar between each GRN population, with the exception of the fru+ male-sensing GRNs. This population was closely associated with the fru+ female-sensing GRNs, more so than were any other 2 GRN subtypes, and the interface between them in UMAP space contained several cells bearing intermediate characteristics. Consequently, the discrepancy in cell numbers between fru+ GRN populations may reflect classification errors due to transcriptomic similarities. (B-F) The UMAP shown in (A) overlaid with the expression of 5 transcription factors (pros, acj6, nvy, fkh, and fru) that are expressed in unique combinations in each of the 4 GRN clusters. (G-AA) Testing the GRN transcription factor code derived from the scRNA-seq data on 24 h APF male first tarsal segments. (G-I) anti-Nvy (green) and anti-Pros (magenta). Note that 1 Pros+ cell is partially obscuring the Pros+ sheath cell, which has a distinct, elongated morphology. (J-L) Anti-Pros (green) and anti-Fkh (magenta). (M-O) Anti-Pros (green) and fru-GAL4 > UAS-mCherry.nls (magenta). (P-R) Anti-Acj6 (green) and fru-GAL4 > UAS-mCherry.nls (magenta). (S-U) Anti-Acj6 (green) and anti-Fkh (magenta). (V-X) Anti-Nvy (green) and anti-Acj6 (magenta). (Y-AA) Anti-Nvy (green) and anti-Fkh (magenta). (AB) A schematic summarizing the expression patterns of each transcription factor across GRNs, along with a selection of other genes detected in each subtype. Gene names are colored pink, green, or blue when shared across multiple GRN subtypes. (AC-AH) Recovery of the same transcription factor code in the Fly Cell Atlas single-nuclei adult male leg neuron data. Note that in the adult data, nvy is barely detected. Correspondence between the nvy+ cluster in the pupal and adult data was supported by additional marker genes, such as foxo and Fer1 (see S11B, S11C, S11H, and S11I Fig). (AC) As in (A), the number of cells in each GRN population is presented. In this dataset, a subregion of what unsupervised clustering labeled as the fru+/acj6− population showed acj6 expression, suggestive of a classification error. This conclusion is further supported by the VGlut, ppk25, and ppk10 data below (see S10Y–S10AB Fig). We therefore manually labeled these as part of the fru+/acj6+ cluster. As in the pupal data, the interface between the 2 fru+ populations appeared particularly close. (AI) A dot plot summarizing the expression of a selection of top differentially expressed genes for each cluster that we identified in the Fly Cell Atlas single-nuclei adult male leg neuron data. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g007
Male- and female-sensing GRNs express distinct receptor repertoires
Previous work has shown that there are 2 fru+ GRNs per leg chemosensory bristle [21,22]. Both express the ion channels ppk23 and ppk29, while one additionally expresses ppk25 and VGlut [21,22,123–126]. These ppk25+/VGlut+ neurons respond to female pheromones, while the ppk25−/VGlut− cells respond to male pheromones [22]. In the pupal data, we observe ppk23 in both fru+ populations, with a small number of ppk29+ cells also distributed across both (S10U and S10V Fig). Although ppk25 was absent in our dataset, we observed a sharp divide between fru+/acj6+ and fru+/acj6- cells based on VGlut expression (S10W Fig). VGlut is restricted to the fru+/acj6− population, identifying these as the female-sensing neurons and the fru+/acj6+ cells as male-sensing. Receptor expression was more readily detected in the adult nuclei data, suggesting that receptors are generally expressed later in development than the time points we sequenced. In the adult dataset, we again observed that VGlut was restricted to the fru+/acj6− population and recovered the previously documented expression patterns of ppk23, ppk25, and ppk29 (S10X–S10AA Fig). However, we also observed GRN-specific expression patterns of several other ppk genes. While ppk25 was restricted to female-sensing cells, ppk10 and the less frequently detected ppk15 were restricted to male-sensing cells (S10AB–S10AD Fig). These therefore represent candidate male pheromone receptors. Conversely, Ir21a and CG46448 appeared to be largely restricted to female-sensing neurons (S10AE–S10AF Fig). CG46448 is adjacent to VGlut on chromosome 2L, so their apparent coexpression in female-sensing neurons points to the possibility of their transcriptional control by shared cis-regulatory elements. These genes aside, the expression differences we observed between male- and female-sensing neurons were generally limited, but several genes were common to both pheromone-sensing populations, including the transcription factors tup and svp, the predicted sulfuric ester hydrolase CG8646, CG14007, and the glycine transporter GlyT, the latter suggesting that these neurons are glycinergic [127] (S10AG–S10AK Fig).
nvy+ GRNs correspond to a sexually dimorphic population required for normal mating behaviour
The nvy+ GRNs showed specific expression of a further set of transcription factors in the pupal data, namely Fer1, foxo, and CG7786 (S11A–S11D Fig). At these time points, few other genes showed enrichment in the nvy+ GRN cluster—exceptions include the phosphodiesterase Pde6 and G-protein–coupled receptor TrissinR (S11E and S11F Fig). To further probe the identity of these neurons, we turned to the adult dataset. The transcription factor code identified in the pupal data largely persisted in the adult data, although each was detected in fewer cells and CG7786 was absent (Figs 7AF and S11G–S11J). AkhR and Gr64a were among the top markers of the nvy+ GRNs in the adult data (S11M and S11N Fig). But like the other 5 Gr genes we detected across the 2 datasets, Gr64a was present in only a handful of cells (S11O–S11T Fig). The sparse detection of Grs may stem from gene dropout due to low abundance or reflect their restricted expression among GRNs of the same class. Ir52a, Ir52b, Ir52c, and Ir52d were also among the top markers of the nvy+ GRN cluster (S11U–S11X Fig). With the exception of Ir52b, these ionotropic receptor (IR) genes have been well characterized: They are known to be largely coexpressed in a subset of leg taste bristles enriched in the first tarsal segment, to show quantitative differences in expression between males and females, to show sexual dimorphism in their projections (they cross the midline in a commissure in males), to be required for normal sexual behavior, and to be expressed in neurons distinct from those involved in sweet or bitter sensing [20,128]. Ir52c and Ir52d are further known to be restricted to the forelegs [20]. Despite their sexually dimorphic characteristics, the neurons expressing these receptors are mutually exclusive from those expressing fru-LEXA within the same bristle [20]. This accords with the scRNA-seq data, where fru appeared largely restricted to the male- and female-sensing populations (Fig 7F and 7AH). Sexual dimorphism in these neurons is therefore instead likely driven by dsx; indeed, at least some Ir52c+ neurons have been shown to descend from a dsx+ lineage [20]. In the adult dataset, dsx expression was patchy among GRNs, appearing enriched in the male- and female-sensing populations (S11Y Fig). In the pupal data, dsx was widely detected across all GRN clusters (S11Z Fig). The discrepancy in the extent of dsx expression in GRNs between the datasets may reflect the differences in the dissected regions: While most male and female pheromone-sensing GRNs may be dsx+ regardless of position, it may be that dsx expression is restricted among nvy+ and fkh+ neurons to those in the regions of the foreleg more likely to contact a mate than food—i.e., the foreleg upper tarsal segments. Restriction of dsx expression to a subset of neurons within a GRN class would provide a mechanism through which an additional layer of between-bristle variation in activity could be achieved.
Distinct and shared modules of gene expression in nvy+ and fkh+ GRNs
The 3 GRN populations that we have discussed—male-sensing, female-sensing, and nvy+—match known populations in the literature. However, we were unable to find mention of a population that resembled our fourth, which was pros+/acj6+/fkh+. Although none of the top 20 DEGs obtained from a comparison with the 3 other GRN populations showed specific expression in the pupal data, there were intriguing similarities with the nvy+ cluster: CAH2, jus, and Glut4EF each looked specific to or highly enriched in both the nvy+ and fkh+ GRNs (S11AA–S11AC Fig). But in the adult dataset, these specific differences were reduced or lost, suggesting that the variation we observed between GRNs in the pupal data may reflect heterochronic differences or that these particular between-GRN developmental differences are lost in adulthood (S11AD–S11AF Fig). Nonetheless, the adult dataset presented its own similarities between fkh+ and nvy+ GRNs: Expression of Ir76b, which is known to be widely expressed among olfactory and gustatory receptor neurons and is thought to form heteromeric complexes with more selectively expressed Ir’s, was restricted to fkh+ and nvy+ GRNs (S11AG Fig) [20,129–131]. The restricted expression of Ir76b contrasts with that of another such coreceptor, Ir25a, which we found broadly expressed across all 4 GRNs (S11AH Fig). Although detected in fewer cells, Ir40a, which is known to be coexpressed with Ir25a in the antennal sacculus, was similarly broadly expressed [132] (S11AI Fig).
The fkh+ population also had something in common with the female-sensing GRNs: Across both datasets, the extracellular matrix proteoglycan gene trol—which we also observed in tendon cells (S6S–S6V Fig)—was enriched in fkh+ and female-sensing GRNs, showed patchy, low-level expression in male-sensing GRNs, and was essentially absent from nvy+ GRNs (S12A–S12D Fig). We recovered this pattern in trol-GAL4 > UAS-mCD8::GFP [133] first tarsal segments counterstained with anti-Pros: Of the Pros+ cells in a single chemosensory bristle, at least 2 were strongly trol-GAL4+ and at least 2 were trol-GAL4− (including the sheath) (S12E–S12G Fig). In some bristles, the remaining Pros+ cell was trol-GAL4+ and in others trol-GAL4−. trol performs several roles during the assembly of the nervous system [134,135]—why it should be limited in its expression among GRNs is unclear. Alongside these shared modules of gene expression, we identified several uniquely expressed genes in fkh+ GRNs, including Tbh, which encodes the key limiting enzyme in octopamine synthesis and therefore suggests that these neurons are octopaminergic (S11AJ and S11AK Fig) [136]. Although very sparsely detected, we also observed Ir60a to be limited to fkh+ neurons (S11AL Fig).
Potential heterochrony in the gene expression profiles of sensory organ neurons
In attempting to identify subtype-specific genes among subclasses of neurons, we observed that genes that appeared cluster-specific in the pupal stages sometimes showed widespread expression in the adult nuclei data. For example, DCX-EMAP, a gene that has been implicated in mechanotransduction in both campaniform sensilla and the chordotonal receptors of the Johnston’s organ [137], switches from being exclusive to campaniform sensilla neurons in the pupal data, to being widespread among both campaniform sensilla and mechanosensory neurons in the adult data (S13C and S13G Fig). Several other genes, such as nAChRalpha7, Ccn, CG1090, CG17839, and CG34370, showed a similar pattern (S13A–S13N Fig). Analogously, MSNCBs and sex comb neurons, identifiable as a vvl- and vvl+/fru+/rn+ subpopulation of mechanosensory neurons, respectively, appeared to develop ahead of the major body of mechanosensory neurons that innervate mechanosensory bristles, as evidenced by the expression patterns across datasets of genes such as sosie, CG31221, and dpr13 (S13O–S13T Fig). In these cases, the broadening of expression between pupal and adult datasets is suggestive of heterochronic differences (i.e., a difference in rate, timing, or duration) in development between the neurons of different sensory organ classes, such that the neurons in mechanosensory bristles lag behind those in chemosensory bristles, campaniform sensilla, and the sex comb. This hypothesis is consistent with documented between-organ variation in developmental timing: Chemosensory bristles are known to be specified earlier than all but the largest mechanosensory bristles [115,116].
Mechanotransduction neurons from different external sensory organ classes express largely shared gene repertoires
Heterochronic differences between sensory organs complicate the identification of organ-specific genes in the pupal data: Genes that appear unique at one stage may be widespread at a later time point. For that reason, we initially focused on the adult nuclei dataset to identify genes enriched in specific non-GRN neuron populations (Fig 7AI). The majority of the top mechanosensory neuron markers that were returned from a DGE analysis comparing these cells to all other neurons appeared nonspecific, being expressed in one or more additional clusters (e.g., Calx, Fife, Dop2R, KrT95D, CG4577, and Ten-m; S14A–S4M Fig). The same applied to the top campaniform sensilla markers, but in this case, despite their lack of complete specificity, many showed a relatively restricted expression profile that extended across both campaniform sensilla and chordotonal organs (e.g., dati, unc79, CG42458, TyrR, Cngl, CARPB, and beat-VI; S14N–S14Z Fig). This pattern is suggestive of certain molecular commonalities between campaniform sensilla and chordotonal organ neurons, commonalities that make them distinct from other mechanotransduction neurons. In further pursuit of genes specific to each of mechanosensory neurons and campaniform sensilla, we tried another approach: identifying top markers of each of these clusters in the pupal data and mapping their expression in adults to determine whether their expression remains cluster specific. Of these, the transcription factor Ets65A, which in the pupal data was restricted to mechanosensory neurons, sex comb neurons, MSNCBs, and campaniform sensilla neurons, remained restricted to mechanosensory neurons and campaniform sensilla neurons in the adult data (S14AA and S14AB Fig). Ets65A therefore represents a candidate regulator of mechanosensory identity in external sensory organ neurons.
We applied this same pupal identification and adult mapping approach in sex comb neurons and MSNCBs, populations that didn’t form distinct clusters in the adult data. No individual gene showed clearly restricted expression to either, but there was strong enrichment of genes that were otherwise patchily expressed across cells. shakB was strongly enriched in sex comb neurons relative to other mechanosensory bristles in the pupal data and widely present across putative chordotonal and campaniform sensilla neurons in the adult data (S14AC and S14AD Fig). In MSNCBs, CG42566, and to a lesser extent CG33639, appeared enriched relative to other mechanosensory populations, with both also detected across GRNs, consistent with a chemosensory bristle origin (S14AE–S14AH Fig). Collectively, MSNCBs and sex comb neurons showed clear enrichment of both effectors of the sex determination pathway: fru and dsx (S14AI–S14AL Fig). The specificity of this enrichment was greater in the case of dsx, which across both neuronal datasets was largely absent outside of the comb, GRN, and MSNCB clusters; fru was surprisingly widespread in the adult data, including expression in the putative chordotonal clusters and campaniform sensilla. The expression of these 2 transcription factors provides a clear regulatory mechanism through which the transcriptomic profiles and activity of these derived mechanosensory populations could readily diverge from other mechanosensory bristles—and do so in a sex-specific manner. But ultimately, determining whether any of the transcriptomic differences between the mechanotransduction neurons that innervate each of mechanosensory bristles, chemosensory bristles, the sex comb, and campaniform sensilla translate into functional differences in operation or sensitivity requires further work. It’s clear from these data that some differences between mechanotransduction neurons innervating different organ classes are present, but they appear minor and on the whole less clear cut than between GRN populations.
Putative chordotonal organ neuron subtypes express specific gene repertoires
In the FCA adult data, we recovered 3 putative chordotonal organ neuron clusters that were absent from our first tarsal segment dataset because these organs fall outside the dissected area. We refer to the largest as “putative chordotonal” and the 2 smaller clusters as erm+ and bab1+ putative chordotonal based on a transcription factor they were each specifically enriched for. Many of the genes we identified as specifically enriched in the putative chordotonal organs in our DGE analysis were absent from the pupal dataset, consistent both with their organ specificity and the absence of chordotonal organs from our dataset. Along with the genes shared among campaniform sensilla neurons and chordotonal organ neurons discussed in the previous section (S14N–S14Z Fig), we identified genes specific to or highly enriched in all chordotonal populations (S15A–S15K Fig), as well as those specific to subpopulations (S15–S15V Fig). The transcriptomic distinctiveness we detect between putative chordotonal neuron clusters aligns with previous work that has identified multiple, functionally distinct neurons in a single chordotonal organ (e.g., “Type A” and “Type B” neurons;, [4,5]). The next step will be to match the distinct clusters we recover to these different chordotonal neuron classes. In turn, that would raise secondary questions, such as whether each neuron class is present in each of the several chordotonal organs that are housed within the leg.
The support cells within a mechanosensory organ each express a distinct gene repertoire
Campaniform sensilla, chemosensory bristles, and mechanosensory bristles each consist of 4 distinct cell types generated through asymmetric divisions of a sensory organ precursor (SOP). The first division gives rise to a progenitor of the socket (tormogen) and shaft (trichogen) cells, while the other to a progenitor of the sheath (thecogen) and neuron(s) (reviewed in [138]) (Fig 8A). Few marker genes are known for the nonneuronal SOP descendants and, to the best of our knowledge, none that definitively separate the same cell type between different organs (e.g., mechanosensory versus chemosensory sockets) [26]. Those markers that are known include the following: Su(H) and Sox15, which specifically accumulate in socket cell nuclei [139,140]; sv (Pax2), which although initially expressed in all bristle cells during the mitotic phase of development is eventually restricted to the shaft and sheath [141]; nompA, which is specifically expressed in sheath cells where it is required to connect dendrites to the shaft [142]; and pros, which is expressed in sheath cells (Fig 6K–6P; [113]).
[Figure omitted. See PDF.]
Fig 8. Distinct and shared modules of gene expression between sensory organ support cells.
(A) The 4 constituent cell types of external sensory organs, such as the mechanosensory bristle in this schematic, originate through asymmetric divisions of a sensory organ precursor (SOP) cell (reviewed in [138]). The SOP divides to produce a pIIa and pIIb daughter cell. pIIa further divides to generate a socket and shaft cell. In the notum, where it’s been studied, pIIb divides into a pIIIb cell and glial cell, the latter of which enters apoptosis soon after birth [188]. pIIIb further divides to produce the sheath and neuron. To the best of our knowledge, whether the pIIb glial division occurs in the leg remains untested. (B) Annotated UMAP plot of the bristle cells from the integrated 24 h AFP and 30 h APF first tarsal segment dataset. The campaniform support cluster included Su(H)+ cells, which suggests that it corresponds to socket cells, but it’s possible that it includes a mix of campaniform sensilla accessory cell types. (C-F) The UMAP shown in (B) overlaid with the expression of a series of marker genes, either previously published or demonstrated in this study, for different sensilla classes or accessory cell types. (G) A dot plot summarizing the expression patterns of a selection of genes identified as being differentially expressed in each of the clusters given in the UMAP shown in (B). Dotted lines separate the 3 major classes of sensory support cell. Of the socket markers, CG31676 is known to be expressed in a subset of olfactory projection neurons [133]; nw is a C-type lectin-like gene; stan, a cadherin that controls planar cell polarity [189]; and nrm, ed, and hbs are cell adhesion molecule genes [143–147]. Of the shaft markers, CG9095 encodes a C-type lectin-like gene; disco-r encodes a transcription factor; dUTPase encodes a nucleoside triphosphate; spdo encodes a transmembrane domain containing protein that regulates Notch signaling during asymmetric cell division [190–192]; and sha encodes a protein involved in the formation of bristle hairs [148]. Aside from pros and nompA, the top markers of the sheaths include the following: the transcription factors Glut4EF, pnt, and SoxN; jv, which encodes a protein involved in actin organization during bristle growth [149]; qua, which encodes an F-actin cross-linking protein [150]; and the midline glia marker wrapper, which encodes a protein involved in axon ensheathment [99–101]. (H-S) The UMAP shown in (B) overlaid with the expression of genes identified in this study as markers of sensory organ support cell subtypes. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.g008
Based on these markers, we identified major shaft, socket, and sheath clusters in our data (Fig 8B–8F). The large size of each of these relative to other clusters in the support cell dataset indicates that they belong to the dominant sensory organ in the first tarsal segment: mechanosensory bristles. Further evidence for a mechanosensory origin comes from the observation that each of these clusters was vvl+, which we previously observed to be expressed in all mechanosensory, but no chemosensory, bristle cells (Fig 6Q–6S). Among the DEGs we identified for each of the mechanosensory shaft, socket, and sheath clusters were many that reflected the biology of these sensory support cells (Fig 8G–8M): the cell adhesion molecule genes nrm, ed, and hbs [143–147] in socket cells; sha, which encodes a protein involved in the formation of bristle hairs [148] in shaft cells; and, in the sheath, a duo of genes, qua and jv, involved in the organization of actin during bristle growth [149,150]. Another nod to the biology of the sheath came in its enrichment for wrapper, which encodes a protein known to be involved in axon ensheathment [101]. Classically used as a marker of midline glia [99–101], the expression of wrapper in sheaths reinforces their glia-like properties, despite not expressing repo or gcm. Moreover, it points to general, shared elements in the mechanisms of ensheathment of neuronal processes between these cell types. The enriched expression in sheaths of a trio of transcription factors—Glut4EF, pnt, and SoxN, of which the latter was completely restricted to sheaths in our dataset—provide a potential route to regulatory divergence from other support cells and glia.
Homologous support cells show transcriptomic divergence between sensory organ classes
We observed that homologous support cells in different sensory organ classes express both shared and distinct gene repertoires. The top markers for each of the mechanosensory socket, shaft, and sheath clusters were enriched in a set of smaller clusters: 3 clusters showed socket-like profiles, 2 shaft-like profiles, and 1 a sheath-like profile (Fig 8G). These minor clusters therefore appear to be sensory organ cell subtypes from sensilla classes that are less abundant in the first tarsal segment than are mechanosensory bristles. Our visualization of eyg-GAL4 > UAS-GFP revealed eyg-GAL4 expression in all 4 cells in a campaniform sensillum (Fig 6AA), so the presence of eyg and toe in the smallest cluster suggest that these cells correspond to a campaniform sensilla population. This cluster uniquely expressed the transcription factor mirr (Fig 8N) and the CPLCP cuticle protein family gene Vajk4, and was Su(H)+, suggesting a socket cell identity, but no further eyg+/toe+ clusters were present. Several explanations for why are plausible: (a) this cluster contains a mix of all campaniform sensilla support cells; (b) only the sockets are transcriptomically distinct enough to cluster separately; and (c) only the sockets were recovered in sufficient numbers to cluster separately. That a small, coclustering group of eyg+ sheath cells were present in the mechanosensory sheath population, rather than forming their own distinct cluster, provides some support for (b) and (c) (Fig 8D). Ultimately, the presence of eyg and toe across the constituent cells of campaniform sensilla suggest that these genes may be master regulators of campaniform sensillum identity.
The minor sheath population was enriched for CG42566, a gene we previously found to be specific to MSNCBs and GRNs among the neurons (S14AE and S14AF Fig), suggesting that this cluster represents descendant cells of the chemosensory pIIb lineage and, therefore, the chemosensory sheath cells specifically. As well as several poorly characterized genes, this cluster showed enriched or unique expression of the lipid-binding protein encoding gene NLaz (Fig 8O), the chitin-binding protein encoding gene Gasp, and Side-VIII. As we observed with neurons, at least some of the differences between mechanosensory and chemosensory sheaths appear to be heterochronic, with chemosensory sheaths developing ahead of their mechanosensory homologs: Several genes, including nompA and wrapper, were widely detected among chemosensory sheath cells, but in mechanosensory sheaths showed localized expression in a region enriched for cells from the 30 h dataset (S16A–S16J Fig).
Based on the expression of Su(H), Sox15, and sv, the remaining clusters have apparent socket and shaft identities. Given the representation of different sensilla classes in the first tarsal segment, the most likely classification for the vvl− clusters are chemosensory sockets and shafts. Except for the extracellular protease gene AdamTS-B, which was heavily enriched in both clusters (Fig 8P), there was no clear transcriptomic link between them. Among the top markers for each, the sv+ putative shaft cluster showed strong enrichment for mtg, which encodes a chitin binding domain-containing protein that’s required to drive postsynaptic assembly [151], while the Su(H)+/Sox15+ putative socket cluster showed unique expression of CG43394 (S17 Fig) and Ance-3 (Fig 8Q), which encodes a predicted membrane component orthologous to human ACE2, the receptor for SARS-CoV-2 [152].
Sex comb cells are enriched for the expression of melanogenic pathway genes
Unlike the chemosensory support cells, the remaining minor putative shaft and socket clusters shared many of their top markers with one another. These genes included the following: b (Fig 8R) and Pxd (Fig 8S), components of the melanogenic pathway [153,154]; dsx, the effector of the sex determination pathway (reviewed in [64]); and fd96Ca, a gene known to contribute to sex comb formation [155] and which we found to be one of the top markers of sex comb neurons in the pupal data. These 2 clusters therefore likely correspond to sex comb shafts and sockets. Whether the genes we find enriched in sex comb sockets and shafts (Fig 8G) are unique to these cells is hard to determine from these data alone due to heterochronic differences between mechanosensory bristles and the sex comb, the latter of which develops earlier. This difference is reflected in the expression of vvl, which was substantially reduced in the putative sex comb shaft cluster relative to both the sex comb socket and the mechanosensory populations (Fig 8E and 8G). Previous work has shown that while Vvl is present in all 4 SOP-descendent cells in external sense organs on the head and notum at 24 h APF, by approximately 42 h, it’s restricted to the socket [156]. Despite the heterochronic differences that likely exist, the enrichment of melanogenic pathway genes is consistent with the sex comb’s heavily melanized appearance. Further work is required to determine whether the changes to mechanosensory bristle cells that generate the modified sex comb morphology operate primarily through quantitative, qualitative, or heterochronic changes in gene expression.
Discussion
Sensory perception begins with contact between a stimulus and a specialized sensory organ. Within and between animal species, these organs are highly variable in their form, cellular composition, and molecular characteristics. To understand the genetic underpinnings of this diversity, both in terms of the developmental networks that specify organ development and the molecular profiles that define mature organs, we characterized the transcriptomes of cells in a region of the male Drosophila foreleg that carries a structurally and functionally diverse selection of sensory organs.
Hierarchical and combinatorial transcription factor codes for leg sensory neurons
The discrete identities adopted by cell types depend on cell type–specific expression of transcription factors, which function as regulators of downstream networks of gene expression. We identified a combinatorial transcription factor code unique to each sensory neuron population present in the first tarsal segment (Fig 9). The “first order” differences in transcription factor expression defined neurons involved in mechanosensation (i.e., those innervating mechanosensory bristles, the sex comb, and campaniform sensilla, along with the MSNCB), which expressed Ets65A, and neurons involved in chemosensation, which expressed pros. The “second order” differences defined subtypes within each of these major classes: The expression of eyg and toe separated campaniform sensilla from other mechanotransduction neurons; acj6, fru, nvy, and fkh delineated GRN subtypes; and vvl separated the mechanosensory neurons of mechanotransduction organs from those that innervate chemosensory bristles. Of these transcription factors, acj6, and to a lesser extent fkh, have repeatedly cropped up in studies of Drosophila neurons, including in subpopulations within the visual, auditory, and olfactory systems [30,47,157–160]. The expression of these transcription factors in a subset of functionally varied neurons points to shared regulatory architecture between them and, by extension, perhaps to common downstream networks of gene expression.
[Figure omitted. See PDF.]
Fig 9. First and second order differences in transcription factor expression between sensory neuron classes.
GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle.
https://doi.org/10.1371/journal.pbio.3002148.g009
Orthogonal regulatory input from the sex differentiation pathway?
The effectors of the sex determination pathway, fru and dsx, showed restricted expression across neuron populations. Two GRNs expressed fru, as did a subpopulation of mechanosensory neurons innervating the sex comb. dsx, on the other hand, was expressed in all GRN populations, sex comb neurons, and in the subpopulation of mechanosensory neurons that innervate chemosensory bristles (MSNCBs). However, the prevalence of fru and dsx expression differed among neuron classes. fru was widely detected in female- and male-sensing GRNs and the sex comb neurons but was only present in a few fkh+ and nvy+ GRNs. In the pupal first tarsal segment data, dsx was detected across all GRNs, although more patchily in fkh+ neurons than in the other subtypes. In the adult GRNs, dsx was more widely detected among female- and male-sensing GRN clusters with a very small number of dsx+ cells present in the remaining GRN populations. The patchiness of expression may reflect gene dropout, with differences in patchiness between populations stemming from quantitative differences in expression. Another explanation, however, is that in some neuron classes, fru and dsx expression is limited to subsets of cells. In this case, sexual identity would serve as a regulatory input orthogonal to the neuron’s core genetic identity and, by extension, provide a regulatory route to sexually dimorphic gene expression in a subset of neurons within a shared class. The orthogonal nature of this regulatory input is most likely to manifest itself in the dsx+ or fru+ subsets of fkh+ and nvy+ GRNs, where dsx and fru expression appears to be relatively rare. In contrast, in the male- and female-pheromone sensing GRNs or the sex comb, fru expression may form a core part of that cell population’s identity.
Reconciling the functional diversity of GRNs with their limited transcriptomic diversity
Considerable diversity is known to exist among leg chemosensory taste bristles in the sensitivities and responsiveness they show to a wide range of tastants, as well as in the identities of the receptors they express [3,19,20,125]. But in all of the datasets we analyzed here—two from a highly localized region of the developing foreleg and one from the entire length of each of the 3 pairs of adult legs—we resolved only the same 4 GRN populations. How, then, can we reconcile the functional diversity known to exist between leg taste bristles with the limited number of transcriptomically distinct GRN classes that we identified? There are several possible solutions. First, a member of each GRN class might not be present in every bristle on the leg, but rather members of a class may be restricted to subsets of bristles. As each chemosensory bristle on the tarsal segments is thought to be innervated by 4 GRNs [16], this would require that some bristles are innervated by multiple GRNs of the same class. Second, it may be that there are additional functionally and transcriptomically distinct GRN classes in the leg, but that they are rare in comparison to the 4 classes we resolved. A very small number of taste bristles at the tips of the leg are known to not be innervated by a ppk25+ female-sensing GRN [125]. This same region also houses some of the least numerous taste bristle classes—classes that are more likely to contact food than potential mates—that have been identified on the basis of morphology and response to tastants [19]. Bristles in this region of the leg would naturally not have made it into our first tarsal segment datasets and their rarity in the wider leg may have limited their recovery in the adult dataset.
There is a third solution to the disconnect between the known functional diversity and transcriptomic similarity that we recover among GRN populations. It might be that the leg taste bristles are largely—or in every case—innervated by a minimal number of 4 functionally distinct GRN classes, each defined by a unique combination of transcription factors. But on top of this core transcriptomic program may be layered an additional level of regulation, one that we were unable to resolve here, which enables the same GRN class in different bristles to express a different receptor or membrane channel repertoire. There is evidence for this in the datasets we looked at here. Ir52c and Ir52d, 2 genes expressed in our nvy+ population, have been shown by both reverse transcription PCR (RT-PCR) and GAL4s to be exclusively expressed in the forelegs, where they are coexpressed [20]. In a dataset composed of all 3 pairs of legs, like the adult nuclei dataset we analyzed, Ir52c+/Ir52d+ cells should therefore comprise a very small number of the total recovered cells, their numbers having been diluted by GRNs from bristles on the other legs. This is indeed what we see, but rather than forming their own cluster, we find the Ir52c+/Ir52d+ cells alongside a greater number that are negative for these 2 genes in a cluster that also includes cells expressing Ir52a, which is known to be expressed in bristles on all legs [20]. Expression patterns such as this, where cells expressing a different set of receptors cocluster, along with the similar representation of cells from each GRN class in our datasets suggest that a “minimal GRN class” model seems likely to underlie most, if not all, taste bristles on the leg.
The nature of the extra regulatory layer that allows receptors to be swapped in and out of a core GRN class remains unclear. The positions of the bristles in which some receptors show restricted expression appear stereotyped [3,19], which suggests that additional layers of regulation may involve positional information. It also suggests that any role for stochasticity in receptor expression, as occurs in mammalian olfactory neurons [161], might be limited. Additionally, restricting the expression of the effectors of sex determination to subsets of bristles would allow for regulatory divergence in neurons from a common GRN class between bristles.
Whatever the differences between the neurons of a single GRN class innervating different bristles may be, our data suggest that the identity of a GRN is more than just the receptors it expresses and that there are deeper transcriptomic features that define a common GRN class. The defining features of a GRN class may relate to the neurotransmitters through which they relay the detection of stimuli or, perhaps, the complement of IR coreceptors they express. On this point, some IRs, including IR25a and IR76b, are known to act as cofactors, forming heteromeric complexes with more selectively expressed IRs [20,129–131]. We found these coreceptors to vary in their specificity, Ir25a being broadly expressed across all 4 GRNs and Ir76b restricted to fkh+ and nvy+ GRNs. We could imagine a modular configuration where coreceptors could set the broad functional range that defines a GRN class—i.e., those classes we recover in our single-cell data—while additional gene products, differentially expressed between neurons of a given GRN class in different bristles, refine GRN function to generate functional diversity between bristles.
Three of the 4 GRN classes we identify match the descriptions of functional populations that have been reported in the literature, each of which is involved in the detection of conspecifics. Two of these are the fru+/ppk23+ “female-pheromone-sensing” and “male-pheromone-sensing” neurons (also known as “F cells” and “M cells”), a pair of neurons that respond selectively to the pheromones of the sex they’re named after [22]. Of the genes we identify as specifically expressed in these cells, ppk10 and ppk15 stand out as candidate male pheromone detectors, given their restriction to male-sensing neurons and the known roles of other ppk genes in pheromone sensing (e.g., [21,123–126]). The other identifiable GRN population that we resolved was the nvy+, Ir52a,c,d-expressing neurons [20]. These neurons are activated by exposure to conspecific females, but not males, and promote courtship. In this sense, naming fru+/ppk23+/ppk25+ GRNs as “female pheromone-sensing neurons”—a name used by others [22] and by us throughout this paper—fails to recognize that these neurons can sit alongside a different GRN with apparently the same broad function in the same bristle. The existence of 2 distinct classes of female-sensing neurons raises the question of why two should be required. That we find ppk genes to be limited to one of the 2 female-sensing GRNs points to differences in either what the neurons respond to or how they respond. A large number of putative pheromones exist in female Drosophila, both in D. melanogaster and in other species [162,163]. This diversity might necessitate multiple detector neurons in order to accurately discriminate between sex and species and perhaps to even allow males to perform finer-grain assessments of the condition or mating status of potential partners. Future work will be required to determine whether responsiveness to these different compounds is divided between the 2 GRN classes, whether there is a logic to that division based on the identity of those compounds, and whether these GRNs project into different neural circuits. Finally, that leaves the 1 GRN population—the fkh+ population—that we couldn’t identify. Whether this class also contributes to conspecific detection and to what extent its function may vary between bristles remains unclear.
The not-so-specific expression of glia markers
Three glia cell types have been described in the Drosophila peripheral nervous system: (1) wrapping glia, which ensheath individual or bundled axons to support the rapid conductance of action potentials; (2) perineural glia, which form the outer cell layer of the nervous system, positioned below a dense network of extracellular matrix called the neural lamella, and which are thought to be responsible for nutrient uptake via the contacts they make with hemolymph; and (3) subperineural glia, large cells that form a thin layer beneath the perineural glia and which establish septate junctions with one another to provide an important structural component of the blood–brain barrier (reviewed in [104,164]). All 3 of these glia are known to be present in the leg [89], and a number of marker genes are known for each [95–97,165,166]. But the glia populations we resolved did not fall neatly along those marker gene lines. moody and, to a lesser extent, Gli, which are both used as specific markers of subperineural glia, were widely detected across our repo+ cells, as were the wrapping glia markers nrv2 and Ntan1 [89,96]. Jupiter, which is thought to specifically mark perineural glia [96], was even more widely detected, being expressed in all nonsensory cells. The noncanonical expression patterns we observe in our glia populations with respect to known marker genes raise difficult questions. Have the GAL4s and protein traps we’ve relied on given us a misleading impression of glia type-specific patterns of gene expression and protein localization? Perhaps—it’s clear that for at least some of these markers antibodies label a wider set of cells than do drivers, as in the case of the apt-GAL4 GMR49G07 [89]. But the surprisingly broad expression of known marker genes is also consistent with an emerging pattern in single-cell studies, where the expression of many genes is considerably wider than expected from reporter and antibody staining data (e.g., [35]). This phenomenon illustrates the importance of cell type–specific posttranscriptional regulation in enforcing the cell type specificity of gene expression, as has been shown for surprisingly widely expressed genes such as the neuronal marker elav [114]. After all, mRNA levels are not the final output of gene expression [167].
A novel cell type and the construction of the neural lamella
We identified a population that didn’t seem to match the description of any reported in the literature. These cells, enriched for the expression of the transcription factors Sox100B and Lim1, exhibited several glia-like properties. For one, they expressed the midline glia marker wrapper [99–101]. More directly, our stainings showed a glia-like association between these cells and both the axon trunks that project from the leg sensory organs to the VNC and the VNC neuropils. Enriched expression of the beaten path genes, beat-IIa and beat-IIb, was further suggestive of a cell that’s in direct communication with others in the developing nervous system [107]. But from a glia perspective, these cells pose a problem: They’re negative for the canonical glia marker repo, a transcription factor expressed in all glia except for those of the midline, and they appear to originate within the leg disc itself, rather than migrating in along with CNS-derived glia. We have 3 clues for what these cells might be. First, a homolog of Sox100B, Sox10, is involved in the specification of glia cells that play similar roles to Drosophila wrapping glia in the vertebrate nervous system (oligodendrocytes and Schwann cells) [168–172]. Indeed, Drosophila Sox100B can rescue vertebrate Schwann cell development in the absence of Sox10 [173]. The failure of Sox100B+ cells to express the wrapping glia marker Oaz would argue against these cells corresponding to wrapping glia, as would their failure to express repo. It may therefore be the case that a conserved developmental program drives wrapping-type morphologies in both these cells and the vertebrate glia subtypes. Which leads us to the second clue: While the expression of membrane-bound GFP revealed narrow cell morphologies when driven by repo-GAL4, under the control of Lim1-GAL4, the staining appeared to encircle a larger area, extending more laterally. The Lim1+/Sox100B+ cells therefore appear to comprise the outer most layer of the nervous tissue. Which leads us to the third clue: These cells were enriched for vkg, a subunit of Collagen IV that’s known to label the neural lamella [96,104]. Collectively, therefore, these data suggest that the Sox100B+ cells are a novel, axon-associated cell type that’s required for the construction of the neural lamella.
Our description of Sox100B+ cells challenges the idea [174] that the source of the neural lamella is the perineural glia (see also [96], who fail to find an effect of RNAi knockdown of vkg on the neural lamella using glia drivers). A more recent alternative hypothesis for the formation of the neural lamella is that its major components—including Collagen IV—are deposited by migrating hemocytes during embryogenesis [96]. This idea is based on the finding that the failure of hemocytes to migrate adjacent to the developing ventral nerve cord is associated with the failure of Collagen IV and another extracellular matrix component, Peroxidasin, to be deposited around it [106]. Two things are interesting to note here. The first is that with the possible exception of NimC3, Sox100B+ cells bear no clear transcriptomic similarity to hemocytes. The second is that hemocytes and Sox100B+ cells show partially overlapping expression of many extracellular matrix component genes. It’s therefore possible that both hemocytes and Sox100B+ cells are collectively required for the construction of the neural lamella during pupal development, each contributing a subset of extracellular matrix components. Resolving the specific role played by Sox100B+ cells in this process, as well as how labor may be divided between additional cell types, represents a key focus for future work.
Materials and methods
Fly strains and husbandry
Flies used in all experiments were raised on a standard cornmeal medium and housed in an incubator at 25°C on a 12:12 cycle. Lines used in this study are detailed in Table 1.
[Figure omitted. See PDF.]
Table 1. Reagents and resources used in this study.
https://doi.org/10.1371/journal.pbio.3002148.t001
Isolation of first tarsal segments for single-cell sequencing
The following procedure was used to collect known-age first tarsal segments for single-cell sequencing. White P1 prepupae (0 to 1 h APF) from the DGRP line RAL-517 [175] were collected and sexed based on the presence of testes. Males were transferred to a folded kimwipe, wet with 500 μl of water, and held inside of a petri dish in an incubator maintained at 25°C on a 12:12 cycle. Pupae were removed from puparia using forceps and placed on top of a water-soaked kimwipe, 1 h before the desired age was reached (e.g., 23 h after collection for the 24 h sample). When the desired age was reached, pupae were placed ventral side up on tape. The base of the abdomen was pierced to release some of the fluid pressure and the foreleg removed at the tibia/tarsal joint. The dissected leg was then placed on tape, covered in a drop of 1X Dulbecco’s PBS (DPBS, Sigma, D8537), and a Micro Knife (Fine Science Tools, 10318–14) was used to sever at approximately the midpoint of the second tarsal segment. The first tarsal segment was then eased out of the pupal cuticle and transferred to a glass well on ice containing 100 μl of 1X DPBS using a BSA-coated 10-μl tip.
Single-cell suspension preparation
We separately prepared 2 single-cell suspensions, each composed of 67 first tarsal segments collected following the procedures described above: one from 24 h APF male pupae, the other from 30 h APF male pupae. For a given time point, once 67 first tarsal segments had been collected, the DPBS was removed from the well and replaced with 100 μl of dissociation buffer, which consisted of 10X TrypLE (Thermo Fisher Scientific, A12177-01) with a final concentration of 2 mg/mL of collagenase (Sigma, C0130). The well was sealed and submerged in a metal bead bath in an incubator at 37°C for 35 min. The dissociation buffer was then removed from the well and replaced with 50 μl of room temperature DPBS before the tissues were subjected to mechanical dissociation. For this, the solution was pipetted up and down 20 times using a 200-μl, widebore, low bind, freshly BSA coated tip (Thermo Fisher Scientific, 2069G) on a 50-μl pipette set to 40 μl. The tip was fully submerged to avoid bringing air into the suspension. The solution was then pipetted up and down a further 20× using a flame-rounded, BSA-coated 200-μl tip, again on a 50-μl pipette set to 40 μl. Next, the solution was pipetted up and down slowly 3 times using the same flame-rounded tip before 40 μl was taken up and transferred to a 2-mL, low-bind, wide-bottomed tube on ice. Exactly 20 μl of DPBS was then slowly dripped around the edges of the well using a different tip to flush the cells into the center. This was then pipetted up and down 3 times using the original flame-rounded 200-μl tip and added to the tube. Cell concentration and viability was assayed using an acridine orange/propidium iodide stain and measured using a LUNA-FL Fluorescent Cell Counter averaged across 2 × 5 μl aliquots (24 h APF sample: 1,990 cells/μl, 98% viability; 30 h APF sample: 967 cells/μl, 98% viability). Through this approach, we were able to avoid filtration or centrifugation steps, both of which we observed to result in high cell loss in trial runs. Although we didn’t perform a controlled test of it, we also observed that the viability of our cell suspensions was considerably higher when using DPBS compared to PBS.
Library preparation and sequencing
Barcoded 3′ single-cell libraries were prepared from single-cell suspensions using the Chromium Next GEM Single Cell 3′ kit v3.1 (10X Genomics, Pleasanton, CA) for sequencing according to the recommendations of the manufacturer. The cDNA and library fragment size distribution were verified on a Bioanalyzer 2100 (Agilent, Santa Clara, CA). The libraries were quantified by fluorometry on a Qubit instrument (LifeTechnologies, Carlsbad, CA) and by qPCR with a Kapa Library Quant kit (Kapa Biosystems-Roche) prior to sequencing. The libraries were sequenced on a NovaSeq 6000 sequencer (Illumina, San Diego, CA) with paired-end 150-bp reads. The sequencing generated approximately 50,000 reads per cell and 500 million reads per library.
scRNA-seq data processing
The alignment, barcode assignment, and UMI counting of the two 10× leg samples were performed using the “count” function in CellRanger (v4.0.0). The reference index was built using the “mkref” function in CellRanger (v3.1.0) and the Ensembl BDGP6.28 Drosophila melanogaster genome. The GTF was filtered to remove non-polyA transcripts that overlap with protein-coding gene models, as recommended in the CellRanger tutorial (https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/using/tutorial_mr). Cell quality filtering and downstream analysis was performed using Seurat (v4.0.5) [176–179] in R (v4.1.1). We began by creating a SeuratObject from the count data using the function “CreateSeuratObject,” excluding cells where fewer than 100 genes were detected. Based on the distribution of the genes and transcripts detected per cell, along with the percentage of reads that mapped to mitochondria, we used the following cell-level filters: 24 h dataset, >450 genes/cell, <5,000 genes/cell, >2,500 transcripts/cell, <10% mitochondrial reads/cell; 30 h dataset, >425 genes/cell, <5,000 genes/cell, >1,400 transcripts/cell, <10% mitochondrial reads. This led to the removal of 1,141 (leaving 9,877) and 1,486 (leaving 10,332) cells from the 24 h dataset and 30 h dataset, respectively. A median of 2,083 genes and 11,292 transcripts were detected per cell in the resulting 24 h dataset, but only 1,245 genes and 5,050 transcripts in the 30 h dataset. This discrepancy is due at least in part to the substantially reduced % of reads mapped confidently to the transcriptome in the 30 h sample (24.0% versus 81.1%), the cause of which we were unable to identify but which did not obviously impact the downstream clustering.
scRNA-seq data clustering
Next, we applied gene-level filtering to the 24 h and 30 h datasets, retaining only those genes expressed in 3 or more cells, and used the function “SCTransform” implemented in Seurat (v4.0.5) to normalize and scale the full 24 h and 30 h datasets [180]. We used 5,000 variable features and regressed out variation due to the percentage of mitochondrial reads. UMAPs of the full datasets were constructed using the “FindNeighbors,” “FindClusters,” and “RunUMAP” functions using principal components (PCs) 1 to 100 and a clustering resolution of 0.7. To identify doublets produced by stochastic encapsulation of multiple cells within a single bead during the microfluidic process, we used the R package DoubletFinder (v2.0.3) [181]. We observed that when run on the full 24 h dataset, DoubletFinder heavily targeted a subregion of the neuron cluster (approximately 29% of all doublets were in the mechanosensory neuron cluster; S18A Fig). Compared to other cells in the neuron cluster, the identified doublets were heavily enriched for the mitotic marker string and cell adhesion gene klingon, the latter of which was restricted to the doublet-enriched region of the mechanosensory neuron cluster as well as a subset of GRNs and mechanosensory sheaths (S18B and S18C Fig). Similar targeting of neurons and sheaths by DoubletFinder was observed in the 30h dataset (S18L Fig). Given this distribution, we reasoned that when run on the entire dataset, DoubletFinder was classifying early differentiating neurons and sheaths, which share a progenitor cell, as doublets. This may be due to their displaying an intermediate transcriptional profile. We therefore ran DoubletFinder separately on the epithelial and nonepithelial datasets. To generate these, we subsetted our data into epithelial cells, identifiable as the large body of contiguous clusters in each dataset (circled in S18A and S18L Fig), and all remaining clusters before reapplying gene-level filtering (retaining those genes expressed in 3 or more cells), rerunning SCTransform (regressing out variation due to the percentage of mitochondrial reads), and reclustering (hereafter all 3 of these processes will be included under the term “reclustering”) (Parameters: 24 h and 30 h datasets: epithelial: PCs = 1:150, variable features = 5,000, r = 0.7; nonepithelial: PCs = 1:50, variable features = 5,000, r = 0.7).
At this stage, the nonepithelial cell datasets contained 2,919 (24 h) and 3,163 (30 h) cells. We then further lowered the mitochondrial percent threshold to <5%, which removed a further 15 (24 h) and 38 (30 h) cells, and nonsheath sensory support cells with >2 nompA transcripts (24 h: 44 cells; 30 h: 96 cells; S18D, S18E, S18M, and S18N Fig). nompA is a known sheath marker [142] and its presence in cells in other sensory support cell clusters indicates possible doublets arising from incomplete dissociation. This left a total of 2,860 (24 h) and 3,029 (30 h) cells, which we reclustered (S18F and S18O Fig) (Parameters: PCs = 1:50, variable features = 5,000, r = 0.7). We then ran DoubletFinder, using the estimated doublet rates outlined in the 10× V3 user guide for the number of cells we recovered (24 h: 8.4%, approximately 11,000 cells; 30 h: 9.3% approximately 11,800 cells; parameters: 1:50 PCs, pN = 0.25, pK = 0.05, without homotypic adjustment). In both datasets, the identified doublets were more evenly dispersed among clusters compared to when run on the full dataset (S18G and S18P). Moreover, genes enriched in the identified doublets were nonspecific and often enriched in epithelial and bract (an identity induced in epithelial cells) clusters (e.g., CG13023) (S18H, S18I, and S18Q Fig), suggestive of them being doublets containing epithelial cells. Cells classified as doublets were removed (217 in 24 h; 259 in 30 h). In the 30 h dataset, a further 9 cells that were enriched for the hemocyte marker NimC4 were removed as presumed doublets from the bract cluster and from the interface between the mechanosensory shaft and socket clusters (S18R Fig). Each dataset was then reclustered (S18J and S18S Fig) (Parameters 24 h: PCs = 1:35, variable features = 5,000, r = 0.7; 30 h: PCs = 1:30, variable features = 5,000, r = 0.7). The 24 h and 30 h nonepithelial datasets were then integrated using the “SelectIntegrationFeatures” (nfeatures = 2,000), “prepSCTIntegration,” “RunPCA,” “FindIntegrationAnchors” (normalization method = SCT, dims = 1:35, reduction = rpca, k anchor = 5), and “IntegrateData” functions in Seurat. Nonsensory cells (clusters enriched for NimC4+, repo+, Sox100B+, aos+, and drm+/vvl+) and sensory support cells (clusters enriched for nompA+, Su(H)+, and sv+) were then separately subsetted and reclustered for downstream analysis (nonsensory: PCs = 1:25, r = 0.4; bristle: PCs = 22, r = 0.2). We identified neurons separately in the nonintegrated datasets based on fne expression, reclustered them (Parameters 24 h and 30 h: PCs = 1:30, var features = 3,000, r = 1.5; 24 h neuron dataset = 514 cells; 30 h neuron dataset = 473 cells; S18K and S18T Fig), and integrated them separately (same parameters as for the integration detailed above but with k anchor of 20).
For epithelial cells, we ran DoubletFinder and separately reclustered the 2 datasets (Parameters 24 h: PCs = 1:150, variable features = 5,000, r = 0.7, with homotypic adjustment, 6,374 cells; 30 h: PCs = 1:30, variable features = 5,000, r = 0.7, with homotypic adjustment, 6,502 cells). We then integrated the 2 datasets using the approach outlined above (Parameters: nfeatures = 2,000, normalization method = SCT, dims = 1:35, reduction = rpca, k anchor = 5), additionally regressing out variation due to cell cycle stage—specifically, the difference between the G2M and S phase scores (calculated following the steps in the Seurat tutorial; https://satijalab.org/seurat/archive/v3.0/cell_cycle_vignette.html). The integrated data were then clustered (PCs = 1:30, r = 0.6), the joints removed and clustered separately (PCs = 10, r = 0.2), and the remaining nonjoint cells clustered (PCs = 20, r = 0.4).
scRNA-seq data analysis
Differential gene expression analyses were conducted on the normalized count data. Data were normalized using the Seurat function “NormalizeData” with normalization method “LogNormalize,” where counts for each cell are divided by the total counts for that cell and multiplied by a scale factor (the default of 10,000) and then natural-log transformed using log1p. To perform the differential gene analysis, we used a Wilcoxon rank sum test implemented through the “FindConservedMarkers” function, grouping cells by their dataset of origin (24 h or 30 h) and specifying that genes were only tested if they were present in >10% of the cells in the focal population. Dot plots and plots of gene expression overlaid on UMAPs were generated using the “DotPlot” and “FeaturePlot” functions, respectively.
Fly Cell Atlas data
To assess the robustness of our developmental neuron classifications in a different, adult dataset, we analyzed the neuronal populations from the FCA leg dataset [38]. This dataset differed in several ways from those we’ve generated here: Ours is single cell, while the FCA data is single nuclei; ours is pupal (24 h APF and 30 h APF), while the FCA data is from adults; ours is from the first tarsal segment of the foreleg, while the FCA data is from the full length of all 3 pairs of legs. We downloaded the “10× stringent” leg loom data (https://cloud.flycellatlas.org/index.php/s/ZX56j2CcMXnHXYc), from which we extracted the gene expression matrix using the “open_loom” and “get_dgem” functions in the “SCopeLoomR” package (https://github.com/aertslab/SCopeLoomR/). We created a SeuratObject using this data, filtering out genes expressed in <3 cells and cells with <100 genes. The dataset was clustered using the functions described above (PCs = 50, r = 1.7) and neurons identified based on fne expression. We then extracted male neurons from this integrated, mixed-sex dataset, which were identifiable from the cell metadata, and reclustered (variable features = 3,000, PCs = 1:25, r = 1).
Fixation, immunohistochemistry, and microscopy
White P1 prepupae (0 to 1 h APF) were collected and sexed under light microscope based on the presence of testes. Males were then placed on a damp kimwipe and aged in an incubator at 25°C until the desired time point. Pupae were then placed on their side on sticky tape and a razor blade used to cut away the dorsal half. The cut pupae were then fixed in a 4% paraformaldehyde solution (125 μl 32% PFA, 675 μl H2O, 200 μl 5X TN) for 50 min on a rotator at room temperature and stored at 4°C. For leg dissections, fixed pupae were removed from the puparia in 1X TNT and tears made in the pupal cuticle at the femur–tibia boundary using forceps. The tibia through to ta5 region was then pulled through the tear, freeing it from the pupal cuticle. At this point, legs expressing fluorescent proteins were mounted in Fluoromount 50 (SouthernBiotech). For immunohistochemistry, the dissected leg region was blocked with 5% goat serum (200 μl 10% goat serum with 200 μl 1X TNT) overnight at 4°C. Legs were then incubated with primary antibody solution overnight at 4°C, washed for 21 min 4 times in 1X TNT, incubated with the secondary antibody solution for 2 h at room temperature, washed for 21 min 4 times in 1X TNT, and then mounted in Fluoromount 50 (SouthernBiotech). All stages, from dissection through to staining, were carried out in a glass well. Antibodies were used in solution with 1X TN and a final concentration of 2% goat serum (see Table 1 for identities and concentrations). Confocal images were taken using an Olympus FV1000 laser scanning confocal microscope or a Zeiss 980 Airyscan. Image stacks were processed using Z-series projection in ImageJ.
Supporting information
S1 Fig. Dataset metrics.
(A) A schematic detailing how many cells were filtered out at each stage of processing. Cells were initially filtered from the full dataset and retained based on the number of genes detected per cell (24 h: >450 and <5,000; 30 h: >425 and <5,000), transcripts detected per cell (24 h: >2,500; 30 h: >1,400), and the percentage of transcripts that map to mitochondrial genes (24 h and 30 h: <10%). The datasets were then split into epithelial and nonepithelial cells based on cluster identity (the asterisks here relate to panels (B-E)). Additional filtering was then performed on the nonepithelial cells, removing cells with >5% mitochondrial reads and nonsheath bristle cells in which >2 transcripts of the sheath marker nompA were detected, which likely correspond to undissociated doublets. Further doublets were then identified in each dataset using DoubletFinder [181] and removed. In the 30 h dataset, an additional 9 cells positive for the hemocyte marker NimC4 were identified at the interface between the mechanosensory socket and shaft cluster and at the edge of the bract cluster. These putative hemocyte–bristle cell doublets were also removed. (B-E) Violin plots showing the distribution of (B) genes detected per cell in the 24 h dataset, (C) genes detected per cell in the 30 h dataset, (D) transcripts detected per cell in the 24 h dataset, and (E) transcripts detected per cell in the 30 h dataset. Panels show the distribution in the full datasets after initial filtering based on cell-level quality control metrics (i.e., the distributions at the positions indicated by asterisks in (A). Numerical data with cell barcodes are listed in S3 Data (24 h) and S4 Data (30 h). Code for generating the figure is available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s001
(PDF)
S2 Fig. Joint markers.
(A) The smallest of the clusters identified in our clustering analysis (Fig 3F) showed highly variable internal expression patterns (i.e., the top markers of this cluster were generally expressed in a relatively small number of its constituent cells). Moreover, this cluster, colored dark green in this plot, showed a markedly higher representation of cells with high mitochondrial read counts, suggesting it may be composed of or enriched for damaged cells. We therefore excluded it from further analysis. Numerical data with cell barcodes are listed in S5 Data. (B-E) UMAP plots of the joint dataset overlaid with the expression of odd-skipped family transcription factors. drm, sob, and odd are known to be expressed in the distal edge of each leg segment except tarsal segments 1–4 [52]. Consistent with its widespread expression among epithelial cells in our dataset, bowl has been shown to display an overlapping but broader expression pattern (extending into tarsal segments 1–4) than odd, drm, and sob [52]. (F, G) UMAP plots of the full joint and nonjoint epithelial dataset overlaid with the expression of bowl (blue) and drm (red; F) or sob (red; G). (H-V) For each panel, a UMAP plot of the joint dataset (left) and full joint and nonjoint epithelial dataset (right) is overlaid with the expression of a given gene identified during the joint differential gene expression analysis. (H, I) CG1648 and Ser show widespread expression among the proximal tibia/ta1 joint and epithelial cells but are excluded from the other joint clusters. This is expected for Ser as Ser+ cells form patterning boundaries in the developing leg that activate joint formation in distally adjacent cells via Notch [193]. (J-N) Lim1, trh, caup, ara, and pdm2 show specific expression in the proximal tibia/ta1 cluster. Of these, Lim1 is known to be expressed in the tibia where it is required for specification of the tarsus ([194]; see also Fig 5V); nub1 alleles give rise to compromised leg development [195]; pdm2 shares cis-regulatory architecture with nub, but, to the best of our knowledge, no role for it in leg development has been characterized [59]; and again, to the best of our knowledge, no roles for the Iro-C genes ara and caup have been reported in leg development. (O, P) Two of the top DEGs for the proximal tibia/ta1 joint, CG2016 and svp, additionally show enriched expression within a common subregion of what we do not label as joint tissue. Given that this region is TfAP-2−, it’s unlikely that this corresponds to the ta2/ta3 joint and instead more likely corresponds to those cells adjacent to the ta1/ta2 joint. (Q, R) Both bab paralogs (bab1 and bab2) are enriched outside of the tibial joint clusters and show particular enrichment within the ta1/ta2 joint cluster, consistent with their known up-regulation in the distal portion of the tarsal segments [56,196]. (S-V) Several of the top DEGs for the ta1/ta2 cluster (CG12880, tnc, CG34938, GstD1) do not appear to be specific, rather they show joint-enriched but widespread expression among epithelial cells. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s002
(PDF)
S3 Fig. Joint and nonjoint epithelial markers.
(A-K) For each panel, a UMAP plot of the joint dataset (left) and full joint and nonjoint epithelial dataset (right) is overlaid with the expression of a given gene identified during the joint cluster differential gene expression analysis. (A-D) Several of the top differentially expressed genes (DEGs) for the distal tibia/ta1 (fj, Sp1, sn, Obp56a) do not appear to be specific, rather they show localized enrichment in joint cells alongside widespread expression in nonjoint epithelial cells. One such gene is fj, which we find widely expressed across all joint and nonjoint clusters but enriched in the distal tibia/ta1 cluster (A; Fig 3I). fj is known to be required for regional growth along the leg’s proximal–distal axis and in imaginal discs shows rings of expression that are complementary to Nub [54,197]. However, our data show a separation between the regions of peak nub and fj expression (compare Fig 3I with Fig 3D). Several of the remaining top DEGs for the ta1/ta2 (E-G) and distal tibia/ta1 (H-K) joint clusters show more specific expression patterns. (L-R) UMAP plots of the joint dataset overlaid with the expression of genes specifying positional identity. Positional identity is clear at the level of the dorsal–ventral axis (ventral: wg, mid, H15; dorsal: dpp, bi; L-P), but not anterior–posterior axis (anterior: ci; posterior: hh; Q, R). (S-AE) UMAP plots of the nonjoint epithelial dataset overlaid with the expression of genes identified as enriched within subregions. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s003
(PDF)
S4 Fig. Pupal leg hemocytes form a uniform population.
(A-W) UMAP plots of the nonsensory dataset overlaid with gene expression. (A-N) A selection of hemocyte markers, most of which are part of a cluster of NIM-repeat containing genes on chromosome 2 that also includes the hemocyte-specific He. RT-PCR work has previously shown that all of these, except nimA, are transcribed in larval hemocytes [69]. We find that all these NIM genes, except nimA, are expressed in our hemocyte cluster, as are Hml and srp, which are known to be expressed in both differentiating and mature plasmatocytes [67,68]. (O-T) Genes identified by Tattikota and colleagues ([70]; see also [72]) as enriched in lamellocytes. We saw no obvious subclustering in relation to these genes: They were either widely expressed among hemocytes (betaTub60D, CG1208), too patchily expressed among hemocytes to reflect a clear subpopulation (atilla, alphaTub85E, CG18754, mthl4), or absent from our dataset entirely (CG31219, CG14610, CG15347, CG12133). (U-W) The same study found that crystal cells showed highest enrichment of PPO1, PPO2, lz, N, peb, and E(spl)m3-HLH. As with lamellocytes, we saw no obvious subclustering in relation to these genes: PPO1, PPO2, and lz were absent from our dataset, and N, peb, and E(spl)m3-HLH showed nonspecific expression. We also mapped the top markers of many of the plasmatocyte subclusters identified by Tattikota and colleagues (Mmp1, IM18, CecA2, CecC, Mtk, DptB, Drs, Prx2540-1, Prx2540-2, CG12896, Abl, Snoo, CG15550, CG6023, mthl7, Cys, CG8860, and COX8), but saw no clear subclustering. (X-AG) Alternating UMAP plots of the full 24 h (X, Z, AB, AD, AF) and 30 h (Y, AA, AC, AE, AG) datasets overlaid with expression of a subset of hemocyte marker genes identified in this study. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s004
(PDF)
S5 Fig. Induction of bract cell identity in epithelial cells is accompanied by a remodeling of the transcriptome.
(A-D) Alternating UMAP plots of the full 24 h (A, C) and 30 h (B, D) datasets overlaid with expression of the bract markers pnt and aos, EGFR signaling components with known roles in bract formation [74–76]. (E) A dot plot of the top 20 differentially expressed genes from a bract versus nonjoint epithelial comparison. Although the comparison was made between all bracts and all nonjoint epithelial cells, the breakdown per subcluster is depicted (“distal” here refers to the sex comb bearing region). Of these, the ecdysone-induced transcription factor Eip93F (also called E93), which we found to be up-regulated in bract cells, is known to be expressed in the epithelial cell that will develop as a bract, where it enables Dll to respond to EGFR signaling [76]. (F) UMAP plot of the full 24 h dataset overlaid with the expression of the top bract marker, CG33110, which encodes a predicted fatty acid elongase. (G) As (F) but in the 30 h dataset, alongside an inset zooming in on the socket cell cluster. The expression of CG33110 in sockets was more pronounced in the 30 h compared to 24 h dataset. (H) A UMAP plot of the nonsensory dataset overlaid with expression of CG10348, a top bract marker. Expression is clearly enriched in bract cells, as well as tendon cells, which are highlighted by a dashed box. (I-P) Alternating UMAP plots of the full 24 h (I, K, M, O) and 30 h (J, L, N, P) datasets overlaid with several top bract markers. In the case of most of these genes, expression is detected outside the bracts and often enriched in the sensory organ cells. Data and code for generating the figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s005
(PDF)
S6 Fig. Characterizing the tendon transcriptome.
(A-R) Alternating UMAP plots of the full 24 h (A, C, E, G, I, K, M, O, Q) and 30 h (B, D, F, H, J, L, N, P, R) datasets overlaid with several top tendon markers. (A-N) For drm, Tsp, CG13003, CG31871, yellow-e, CG13722, and CG9650, expression was observed in both tendon cells and a subregion of epithelial cells. For tx (O, P) and CG42326 (Q, R), we also observed expression in a subset of shaft and socket cells. In both cases, this socket and shaft expression was more widespread at 30 h. (S) Confocal images of 24 h APF male trol-GAL4 > UAS-mCD8::GFP (green) [133] legs counterstained with the neuronal marker anti-Futsch (magenta). The first 3 images show the separate and merged channels from an image of the first tarsal segment. The staining follows much the same pattern showed by anti-Vvl and 1151-GAL4 > UAS-mCherry.nls in the tendon cells (Fig 4G–4J). Note the concentration of staining around the tibia/ta1 joint, the position of the levator and depressor tendons. The right-hand image shows the distal tibia and proximal ta1 with merged channels. Note the presence of extensive trol-GAL4 staining in epithelial cells in the tibia—no equivalent epithelial staining was observed in the tarsus. The epithelial trol-GAL4 staining observed in the tibia was not present in the region proximal to the tibia/ta1 joint. trol-GAL4 staining was also observed in gustatory receptor neurons (GRNs) (see S12 Fig). (T-V) Expression of trol overlaid on the nonsensory (T), full 24 h (U), and full 30 h (V) UMAP plots. Note the expression of trol in a subset of GRNs (as shown in (S) and S12 Fig). Some localized expression is present in a region of the epithelial clusters that corresponds to the proximal tibia/ta1 portion of our joint UMAPs, rather than the distal tibia/ta1 region, and therefore likely reflects the tibia/ta1 joint staining rather than that in the more distal tibia, which falls outside of our dissected region. (W-AE) Alternating UMAPs showing expression of known tendon genes sr, Lrt, and slow overlaid on the nonsensory (W, Z, AC), full 24 h (X, AA, AD), and full 30 h (Y, AB, AE) datasets. Note the enriched expression of sr in a nontendon region of the nonsensory UMAP (W). These cells correspond to the Oaz+/repo+ glia. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s006
(PDF)
S7 Fig. Sox100B+ cells originate within the leg disc in contrast to migratory CNS-derived glia.
(A) A dot plot of the top differentially expressed genes from between-glia comparisons (e.g., Oaz+/repo+ glia vs. svp+/repo+ and Sox100B+ glia, etc.). (B) L2 larval brain, ventral nerve cord (VNC), and leg disc from a repo-GAL4 > UAS-mCherry.nls (magenta) male counterstained with anti-Sox100B (green). An inset is shown with a close-up of the disc. Note the staining running along the anterior–posterior axis of the VNC and extending up into the brain. In the VNC, this staining falls along the boundary of the neuropil (see S7D Fig). (C) As in (B) but an L3 larval leg disc. (D) The posterior end of the VNC from a 0 h after puparium formation (APF) male stained with both anti-Sox100B (green) and anti-Futsch (magenta), a neuronal marker. Note how anti-Sox100B staining hugs the outer boundary of the neuropil. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s007
(PDF)
S8 Fig. Annotation of glia clusters.
(A, B) 24 h APF male first tarsal segments from repo-GAL4 > UAS-mCD8::GFP (green) counterstained with anti-Futsch (magenta). Note how the staining pattern includes cells hugging the axon trunks, but also, and unlike in the Lim1-GAL4 staining, cells that branch away from them. This latter feature is particularly clear around the sex comb (see white triangles). Note also that some circular, nonspecific fat body staining is present (examples marked by asterisks in (A)). (C) 24 h APF male forelegs from 1151-GAL4 > UAS-mCherry.nls (magenta) males counterstained with anti-Mef2 (green). 1151-GAL4 is a myoblast and tendon cell marker, while Mef2 is a transcription factor essential for cardiac, visceral, and somatic muscle development [203]. Anti-Mef2 staining is restricted to the tibia. (D) Confocal image of a male 48 h pupal leg from a vkg::GFP line. Both transmitted light and GFP channels are shown merged. (E) Annotated UMAP plot of nonsensory cells. (F-K) The UMAP shown in (E) overlaid with the expression of: (F) Jupiter, a perineural glia marker [96] that here shows widespread expression; (G) Gs2, a surface glia marker that here shows patchy expression in the svp+/repo+ cluster [89]. (H-K) A series of extracellular matrix components that show varying expression profiles across repo+ glia, Sox100B+ glia, and hemocytes. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s008
(PDF)
S9 Fig. Annotation of sensory neuron populations.
(A-H) In our unsupervised clustering analysis of both the pupal neuron and the FCA neuron datasets, a small subpopulation of mechanosensory neurons clustered separately, labeled in (A) and (E) as TkR86C+ mechanosensory neurons. These clusters were enriched for TkR86C (A, E), which encodes a receptor for the neuropeptide tachykinin and plays a critical role in male-specific neural, circuits that control aggression [198]. (B-D) In the pupal dataset, these cells were also enriched for the ventral marker H15, CARPA, and Side-II. (F-H) However, in the adult data, only CARPA showed any suggestion of being enriched in these cells relative to the other neuron populations. Because of their scarcity, coupled with the absence of strongly specific genes, it’s unclear whether the TkR86C+ cluster represents a distinct population. We cannot rule out that in the pupal data, they may simply correspond to more developmentally advanced mechanosensory neurons and/or a population from a particular subregion of the leg, the clustering of which is driven by the shared expression of positional markers such as H15. (I) A UMAP plot of an integrated dataset of the 24 h APF male first tarsal segment single-cell RNA-seq data, 30 h APF male first tarsal segment single-cell RNA-seq data, and adult all leg male neuron single-nuclei RNA-seq data. Cells are colored according to the dataset of origin. (J) The UMAP plot given in (I) but this time cells are colored according to the cluster annotation they were assigned based on separate clustering and analysis of the pupal cell and adult nuclei datasets (i.e., those presented in Fig 6A and 6G). Note how the nvy+ cluster includes both cells labeled as nvy+ gustatory receptor neurons (GRNs) and mechanosensory neurons. Because of this divergent classification, and the broader differences between the datasets in the tissues, their ages, and the dissociation protocol used, we opted to analyze the 2 datasets separately. (K-M) Confocal images of mechanosensory bristles in a subregion of the first tarsal segment from elav-GAL4 > UAS-mCherry.nls (magenta) males, counterstained with anti-Pros (green). The channels are shown separately and merged. Note the presence of both Pros+/elav-GAL4+ and Pros+/elav-GAL4- cells. This heterogeneity likely reflects between-bristle variation in the developmental stage of mechanosensory sheath cells such that soon after division mechanosensory sheaths are elav-GAL4+ (see also [113]). elav-GAL4 expression is later lost from mechanosensory sheaths, restricting it to neurons. (N) A UMAP plot of the Fly Cell Atlas single-nuclei adult male leg neuron data overlaid with the expression of CG42566. Expression of CG42566 is largely restricted to the 4 gustatory receptor neuron (GRNs) classes. In the pupal data, it is also expressed in a subpopulation of vvl− mechanosensory neurons, which we presume to correspond to the mechanosensory neurons that innervate chemosensory bristles (MSNCBs). It’s possible that the patchy expression of CG42566 in the mechanosensory neuron cluster observed here in the adult data reflects MSNCBs interspersed among mechanosensory neurons. Interestingly, CG42566 is present in nvy+ GRNs in the adult data but absent from them in the pupal data, which may reflect between-GRN developmental timing differences. (O) Confocal images of adult male eyg-GAL4 > UAS-GFP.S65T legs. On the left, the first 2 tarsal segments (ta1 and ta2) are shown; on the right, the second through to the fifth (ta2-ta5). Blue arrows and text denote the name and position of visible, stained campaniform sensilla. The naming follows the nomenclature of Dinges and colleagues [8]. Under this nomenclature, the first 3 characters denote the tarsal segment; the fourth, whether the sensilla are found singly (S) or in a group (G) of 2 (as in distal ta1 and ta3) or more (as in ta5); and the fifth, that they are on the front (F) leg. In Ta1GF, note how the cell body of the neuron sits a little further back from the accessory cells, a position it may adopt due to the rotation of the sex comb. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s009
(PDF)
S10 Fig. Gene expression profiles of gustatory receptor neurons (GRNs) I.
(A-T) UMAPs showing the annotated GRN clusters identified in the integrated pupal neuron data (A) and FCA adult leg data (G) and then overlaid with the expression of genes identified as being specifically expressed or enriched in all GRNs relative to all other neuron populations. (U-AK) The expression of a set of genes that are enriched in the female-sensing and/or male-sensing GRN clusters overlaid on the UMAPs described in (A) and (G). Of these, ppk23 and ppk29 are known from previous work to be expressed in both neuron types, while VGlut is restricted to female-sensing neurons [21,22,123–126]. We additionally detect the transcription factor tup in both populations and ppk10 and ppk15 in the male-sensing population. Note that in (AD), 2 of the 3 ppk15+ cells outside the main body of M-sensing GRN cells fall within the acj6+ region we identified as likely being additional M-sensing cells (see Fig 7AH). Taking the 2 datasets together, ppk15 represents a strong candidate for a gene involved in male pheromone detection, as does ppk10. A gene plotted for just one of the 2 datasets indicates its absence from the other. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s010
(PDF)
S11 Fig. Gene expression profiles of gustatory receptor neurons (GRNs) II.
(A-N) UMAPs showing the annotated GRN clusters identified in the integrated pupal neuron data (A) and FCA adult leg data (G) and then overlaid with the expression of genes identified as being specifically expressed or enriched in the nvy+ GRN cluster. (O-T) As well as searching through DEGs, we ran through the expression of all 60 gustatory receptor genes [19,199–202]. Of these, we detected Gr36a, Gr47b, Gr58a, and Gr63a exclusively in the pupal ta1 dataset, Gr64a exclusively in the adult leg dataset, and Gr61a in both. In all cases, expression was limited to just a handful of cells precluding us from confidently assigning Grs to specific GRNs. We failed to detect Gr68a, a receptor that has been shown to be specifically expressed in the male foreleg [3], in either dataset. (U-X) Ir52a-d, the expression of which is shown here overlaid on the UMAP presented in (G), were specifically expressed in the nvy+ GRN cluster and only detected in the adult dataset. (Y-Z) The expression of dsx appears more widespread among the GRNs in the pupal data compared to the adult data. This may reflect differences in the dissected regions, with dsx expression restricted to only the nvy+ and fkh+ GRNs on the foreleg, while dsx is expressed in all fru+ neurons regardless of the leg they’re on. (AA-AL) A selection of genes enriched specifically in fkh+ GRNs or that are shared between fkh+ and nvy+ GRNs. Note how initially restricted expression in the pupal data of CAH2 (AA), jus (AB), and Glut4EF (AF) expands to widespread expression in the adult data (AD-AF). Genes presented for just one of the 2 UMAPs indicates its absence from the other dataset. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s011
(PDF)
S12 Fig. Variable expression of the extracellular matrix proteoglycan gene trol between gustatory receptor neuron (GRN) classes.
(A) A UMAP showing the annotated GRN clusters identified in the integrated pupal neuron data. (B) The UMAP shown in (A) overlaid with the expression of trol, which encodes the extracellular matrix proteoglycan Perlecan. (C) A UMAP showing the annotated GRN clusters identified in the Fly Cell Atlas single-nuclei adult male leg neuron data. (D) The UMAP shown in (C) overlaid with the expression of trol. (E-G) Confocal images of 30 h APF male first tarsal segments from trol-GAL4 > UAS-mCD8::GFP (green) counterstained with anti-Pros (magenta). We imaged at 30 h APF because at 24 h APF, trol-GAL4 expression in the bristles of the first tarsal segment was either undetectable or very weak. However, at 24 h APF, trol-GAL4 expression was clear in the more distal segments, suggesting these bristles may develop ahead of those in the more proximal segments. The imaged region contains 5 chemosensory bristles, which are circled and numbered in (E). anti-Pros marks 5 nuclei (marked with an asterisk) in each bristle: 4 correspond to GRNs and 1 to the sheath. Individual Pros+ nuclei are visible outside of the circled chemosensory bristles shown in (E). These correspond to the sheath cells of mechanosensory (MS) bristles. In (G), note how there is between-bristle variation in the number of trol-GAL4+ cells. Bristle 1: 2 strongly positive, 1 weakly positive, 2 negative; Bristle 2: 2 positive, 3 negative; Bristle 3: 2 positive, 3 negative; Bristle 4: 3 positive, 2 negative; Bristle 5: 3 positive, 2 negative. This variability matches the expression profile shown in the UMAPs (B, D), where F-sensing and fkh+ GRNs show strong trol expression and M-sensing GRNs show variable expression. Data and code for generating the scRNA-seq elements of this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s012
(PDF)
S13 Fig. Potential heterochronic differences between sensory neuron classes.
(A) Annotated UMAP of the pupal integrated neuron data. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. (B) Annotated UMAP of male neuronal cells subsetted from the Fly Cell Atlas single nuclei RNA-seq leg dataset [38]. (C-N) The UMAPs described in (A) and (B) overlaid with a selection of genes identified among the top markers of campaniform sensilla neurons in the pupal data and which show a loss of specificity when moving from the pupal to adult dataset. (O-T) The UMAPs described in (A) and (B) overlaid with a selection of genes identified among the top markers of MSNCBs and/or sex comb neurons in the pupal data and which show a loss of specificity when moving from the pupal to adult dataset. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s013
(PDF)
S14 Fig. Gene expression in external mechanotransduction neurons.
(A) Annotated UMAP of the pupal integrated neuron data. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. (B) Annotated UMAP of male neuronal cells subsetted from the Fly Cell Atlas single-nuclei RNA-seq leg dataset [38]. (C-AJ) The UMAPs described in (A) and (B) overlaid with a selection of genes showing enriched expression in the different external sensory organ neuron classes involved in mechanotransduction, namely mechanosensory neurons, MSNCBs, sex comb neurons, and campaniform sensilla. (A-M) Genes identified as top markers of mechanosensory neurons in the adult FCA data, but all show expression in other populations. (N-Z) Genes identified as top markers of campaniform sensilla neurons in the adult FCA dataset. Note how many are also expressed in chordotonal organ populations, but few or no mechanosensory neuron populations. (AA-AB) Across both datasets, Ets65A appears largely restricted to mechanosensory neurons, MSNCBs, sex comb neurons, and campaniform sensilla. (AC-AD) Although relatively widely expressed in the adult data, shakB show marked enrichment in sex comb neurons in the pupal data. (AE-AH) CG42566 and, to a lesser extent, CG33639 appear enriched in MSNCBs. (AI-AL) The 2 effectors of sexual differentiation, fru and dsx, show distinct expression profiles from one another. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s014
(PDF)
S15 Fig. Gene expression in putative chordotonal organ neurons.
(A) Annotated UMAP of the pupal integrated neuron data. GRN, gustatory receptor neuron; MSNCB, mechanosensory neuron in chemosensory bristle. (B) Annotated UMAP of male neuronal cells subsetted from the Fly Cell Atlas single-nuclei RNA-seq leg dataset [38]. (C-V) UMAPs of the pupal and adult neurons overlaid with the expression of genes identified as being enriched in the putative chordotonal clusters in the adult data. Note that of these, only CG3339, CG9650, CG43689, and yuri were present in the pupal neuron dataset. (C-K) CG9492, CG10185, CG13133, CG9650, nan, CG14342, Hsc70-2, and CG43149 were found across all putative chordotonal organ cells and largely or entirely absent from the pupal ta1 neuron data. (L) Dhc93AB was present in all chordotonal clusters except for the bab1+ population. (M) NPFR was present in all chordotonal clusters except for the erm+ population. (N-P) The population enriched for the expression of the transcription factor erm showed specific expression of CG14693, Dhc98D, and CG13408. (Q, R) Among chordotonal populations, the transcription factor yuri was largely restricted to the erm+ cluster but also showed expression in GRNs. (S-V) The bab1+ population showed enriched expression of CG3339 and CG43689, although both genes showed spatially restricted expression in a subset of cells in the major chordotonal cluster. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s015
(PDF)
S16 Fig. Heterochronic differences between mechanosensory and chemosensory sheath cells.
(A) A subset of the sensory support cell UMAP shown in Fig 8B showing only the mechanosensory and chemosensory sheath populations. Cells are colored in relation to their dataset of origin and therefore, by extension, the time point after puparium formation at which they were collected. (B-J) The UMAP shown in (A) overlaid with the expression of genes in the mechanosensory sheath cluster that we identified as being significantly up-regulated in cells from the 30 h dataset compared to those from the 24 h dataset. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s016
(PDF)
S17 Fig. Visualizing the expression of CG43394.
Confocal images of a 24 h APF male first tarsal segment from CG43394-GAL4 > UAS-mCD8::GFP counterstained with the neuronal marker anti-Futsch. Visible cell bodies are highlighted with arrows in (A). CG43394 was one of the top markers for a cluster we identified as socket cells from chemosensory bristles. This annotation was based on the strong transcriptomic overlap between the cluster and the other socket populations, including the shared expression of the known socket transcription factors Su(H)+ and Sox15+ (Fig 8G). However, the CG43394-GAL4 > UAS-mCD8::GFP staining does not appear to correspond to chemosensory sockets, but rather to a trachea-like channel running through the middle of the tarsal segments. While we cannot exclude the possibility that the cluster we annotated as chemosensory sockets in fact corresponds to these cells, we have reason to doubt that it does. In addition to the socket-like transcriptomic profile of the cells, it seems unlikely that a cell type with approximately 2 cells in the first tarsal segment would generate a cluster of equivalent size to the CG43394+ population. For comparison, this cluster included 69 cells, compared to the 84, 74, and 76 in each of the putative sex comb sockets and shafts and chemosensory sheaths, respectively (approximately 11 of each are found in a single ta1).
https://doi.org/10.1371/journal.pbio.3002148.s017
(PDF)
S18 Fig. Processing the 24 h AFP and 30 h APF male first tarsal segment scRNA-seq datasets.
(A) A UMAP plot of the 24 h dataset after low-quality cells have been removed (i.e., those that fail to meet the criteria of >450 genes/cell, <5,000 genes/cell, >2,500 transcripts/cell, <10% mitochondrial reads/cell). Putative doublets identified by DoubletFinder are colored red and singlets blue. Note how in these data, Doublets are heavily enriched within a subregion of mechanosensory neurons and hemocytes. Approximately 29% of cells in the mechanosensory neuron cluster were labeled as doublets. (B) The UMAP plot shown in (A) overlaid with the expression of the mitotic marker stg. Note how the same region of the mechanosensory neuron cluster that is enriched for “doublets” is also enriched for stg. (C) The UMAP plot shown in (A) overlaid with the expression of klg, a gene we identified as one of the top markers of the “doublets” when compared to other mechanosensory cells in the cluster. The only other regions of the UMAP where klg expression is detected is in closely associated subsets of mechanosensory sheaths and gustatory receptor neurons (GRNs). By itself, this restricted expression pattern suggests that these cells are not bona fide doublets. Rather, and considered alongside the stg expression, it suggests that these cells are early differentiating neurons. Given that neurons and sheaths are formed from the same cell division, it further suggests that klg is an early marker of both cell types. (D) The percentage of reads per cell that map to mitochondrial genes overlaid on a UMAP of all the 24 h nonepithelial cells. Note the presence of high % cells at the interface between the socket and shaft clusters. (E) The expression of the sheath marker nompA overlaid on a UMAP of all the 24 h nonepithelial cells. Note the presence of nompA+ cells in other sensory support cell clusters (shafts and neurons; see pink arrows). The presence of nompA is likely indicative of doublets arising through incomplete dissociation of these tightly associated cells within sensory bristles. “MS” = mechanosensory. (F) A UMAP plot of all 24 h nonepithelial cells after removal of sensory support cells with >2 nompA transcripts detected or where more than 5% of reads mapped to mitochondrial genes. (G) The UMAP shown in (F) with putative doublets identified by DoubletFinder colored red and singlets blue. When run on this subsetted data, the identified doublets are more evenly dispersed among clusters than when run on the full dataset (as shown in A). (H) The UMAP shown in (F) overlaid with the expression of CG13023, one of the top markers of the doublets identified in the neuron cluster. CG13023 is enriched in all areas of the UMAP where doublets were identified as well as in the bract clusters. Bracts develop from epithelial cells, which suggests that the doublets identified are likely to be bona fide doublets that include an epithelial cell. (I) As (H) but overlaid on the full 24 h UMAP shown in (A). CG13023 is enriched in epithelial cells, further supporting the conclusion that the CG13023 enriched cells identified by DoubletFinder are bona fide doublets. (J) A UMAP plot of the 24 h nonepithelial cells clustered after the removal of doublets identified in (G). (K) A UMAP plot of the neurons subsetted from (J) and reclustered. (L) A UMAP plot of the 30 h dataset, post-cell filtering, with putative doublets identified by DoubletFinder colored red and singlets blue. As in the 24 h data, doublets are heavily enriched within a subregion of mechanosensory neurons and hemocytes. (M) A UMAP plot of the subsetted nonepithelial cells overlaid with the percentage of reads per cell that map to mitochondrial genes. Note the presence of high % cells at the interface between the socket and shaft clusters. (N) The expression of the sheath marker nompA overlaid on a UMAP of all the 30 h nonepithelial cells. As in the 24 h dataset, note the presence of nompA+ cells in other sensory support cell clusters (shafts and neurons; see pink arrows). (O) A UMAP plot of all 30 h nonepithelial cells after removal of sensory support cells with >2 nompA transcripts detected or where more than 5% of reads mapped to mitochondrial genes. (P) The UMAP shown in (O) with putative doublets identified by DoubletFinder colored red and singlets blue. As with the 24 h data, when run on the nonepithelial subset of cells, the identified doublets are more evenly dispersed among clusters than when run on the full dataset (as shown in (L)). (Q) As in the 24 h data, cells identified as doublets are enriched for CG13023, a gene that shows high expression in epithelial cells, from which bracts derive. The full 30 h UMAP shows an equivalent pattern to that observed in the full 24 h UMAP in (I). (R) As shown in (P), DoubletFinder identified several doublets at the interface between the mechanosensory shafts and socket clusters, as well as on the periphery of the bracts. Closer inspection of other cells in this region (see arrows) revealed that many of the neighboring cells identified as singlets expressed high levels of the hemocyte marker NimC4, suggesting that they too were doublets. These cells were manually highlighted and removed. (S) A UMAP plot of the 30 h nonepithelial cells clustered after the removal of doublets identified in (P). (T) A UMAP plot of the neurons subsetted from (S) and reclustered. No campaniform sensilla (i.e., eyg+/toe+ cells) neurons are readily detected in this UMAP. Data and code for generating this figure are available at https://www.osf.io/ba8tf.
https://doi.org/10.1371/journal.pbio.3002148.s018
(PDF)
S1 Data. Cell identities and barcodes for Fig 2L.
https://doi.org/10.1371/journal.pbio.3002148.s019
(CSV)
S2 Data. Cell identities and barcodes for Fig 2N.
https://doi.org/10.1371/journal.pbio.3002148.s020
(CSV)
S3 Data. Transcripts and genes detected per cell barcode in the 24 h dataset after initial cell quality filtering.
Data for S1B and S1D Fig.
https://doi.org/10.1371/journal.pbio.3002148.s021
(CSV)
S4 Data. Transcripts and genes detected per cell barcode in the 30 h dataset after initial cell quality filtering.
Data for S1C and S1E Fig.
https://doi.org/10.1371/journal.pbio.3002148.s022
(CSV)
S5 Data. Percentage of reads mapping to mitochondrial genes per cell barcode in the epithelial joint dataset.
Data for S2A Fig.
https://doi.org/10.1371/journal.pbio.3002148.s023
(CSV)
Acknowledgments
We thank Diana Burkart-Waco, Emily Kumimoto, and Hong Qiu for preparing libraries from our single cell suspensions and performing the sequencing; John Larue, Aidan Angus-Henry, and Tiezheng Fan for assistance with collecting, fixing, and mounting pupae; Kohtaro Tanaka for assistance in developing the first tarsal segment dissection technique; Alex Majane, Evan Witt, and Allison Jevitt for answering questions that helped us design our tissue dissociation protocol; Michael Paddy and Thomas Wilkop for microscopy assistance; and Michel Gho for sharing his observations of Pros expression. Thanks also to Liqun Luo, Cedric Soler, Makoto Sato, David Bilder, Hui-Yu Ku, and the Bloomington Stock Center for Drosophila strains, and to the Developmental Studies Hybridoma Bank, Steve Russell, Sarah Certel, Vidya Chandrasekaran, Herbert Jackle, Richard Mann, Marc Freeman, and Megan Corty for antibodies.
Citation: Hopkins BR, Barmina O, Kopp A (2023) A single-cell atlas of the sexually dimorphic Drosophila foreleg and its sensory organs during development. PLoS Biol 21(6): e3002148. https://doi.org/10.1371/journal.pbio.3002148
1. Guo L, Zhang N, Simpson JH. Descending neurons coordinate anterior grooming behavior in Drosophila. Curr Biol. 2022;32:823–833.e4. pmid:35120659
2. Spieth HT. Courtship behavior in Drosophila. Annu Rev Entomol. 1974;19:385–405. pmid:4205689
3. Bray S, Amrein H. A Putative Drosophila Pheromone Receptor Expressed in Male-Specific Taste Neurons Is Required for Efficient Courtship. Neuron. 2003;39:1019–1029. pmid:12971900
4. Mamiya A, Gurung P, Correspondence JCT, Tuthill JC. Neural Coding of Leg Proprioception in Drosophila Article Neural Coding of Leg Proprioception in Drosophila. Neuron. 2018;100:636–650.e6. pmid:30293823
5. McKelvey EGZ, Gyles JP, Michie K, Barquín Pancorbo V, Sober L, Kruszewski LE, et al. Drosophila females receive male substrate-borne signals through specific leg neurons during courtship. Curr Biol. 2021;31:3894–3904.e5. pmid:34174209
6. Zill S, Schmitz J, Büschges A. Load sensing and control of posture and locomotion. Arthropod Struct Dev. 2004;33:273–286. pmid:18089039
7. Akay T, Haehn S, Schmitz J, Büschges A. Signals from load sensors underlie interjoint coordination during stepping movements of the stick insect leg. J Neurophysiol. 2004;92:42–51. pmid:14999042
8. Dinges GF, Chockley AS, Bockemühl T, Ito K, Blanke A, Büschges A. Location and arrangement of campaniform sensilla in Drosophila melanogaster. J Comp Neurol. 2021;529:905–925. pmid:32678470
9. Morphology Hannah-Alava A. and chaetotaxy of the legs of Drosophila melanogaster. J Morphol. 1958;103:281–310.
10. Walker RG, Willingham AT, Zuker CS. A Drosophila mechanosensory transduction channel. Science (80-). 2000;287:2229–2234. pmid:10744543
11. Tanaka K, Barmina O, Kopp A. Distinct developmental mechanisms underlie the evolutionary diversification of Drosophila sex combs. Proc Natl Acad Sci. 2009;106:4764–4769. pmid:19255422
12. Tokunaga C. Cell lineage and differentiation on the male foreleg of Drosophila melanogaster. Dev Biol. 1962;4:489–516. pmid:13921596
13. Ng CS, Kopp A. Sex combs are important for male mating success in Drosophila melanogaster. Behav Genet. 2008;38:195–201. pmid:18213513
14. Massey JH, Chung D, Siwanowicz I, Stern DL, Wittkopp PJ. The yellow gene influences Drosophila male mating success through sex comb melanization. elife. 2019;8:673756. pmid:31612860
15. Stocker RF. The organization of the chemosensory system in Drosophila melanogaster: a rewiew. Cell Tissue Res. 1994;275:3–26. pmid:8118845
16. Nayak SV, Singh RN. Sensilla on the tarsal segments and mouthparts of adult Drosophila melanogaster meigen (Diptera: Drosophilidae). Int J Insect Morphol Embryol. 1983;12:273–291.
17. Vosshall LB, Stocker RF. Molecular architecture of smell and taste in Drosophila. Annu Rev Neurosci. 2007;30:505–533. pmid:17506643
18. Montell C. A taste of the Drosophila gustatory receptors. Curr Opin Neurobiol. 2009;19:345–353. pmid:19660932
19. Ling F, Dahanukar A, Weiss LA, Kwon JY, Carlson JR. The Molecular and Cellular Basis of Taste Coding in the Legs of Drosophila. J Neurosci. 2014;34:7148–7164. pmid:24849350
20. Koh TW, He Z, Gorur-Shandilya S, Menuz K, Larter NK, Stewart S, et al. The Drosophila IR20a Clade of Ionotropic Receptors Are Candidate Taste and Pheromone Receptors. Neuron. 2014;83:850–865. pmid:25123314
21. Thistle R, Cameron P, Ghorayshi A, Dennison L, Scott K. Contact Chemoreceptors Mediate Male-Male Repulsion and Male-Female Attraction during Drosophila Courtship. Cell. 2012;149:1140–1151. pmid:22632976
22. Kallman BR, Kim H, Scott K. Excitation and inhibition onto central courtship neurons biases Drosophila mate choice. elife. 2015:4. pmid:26568316
23. Lai EC, Orgogozo V. A hidden program in Drosophila peripheral neurogenesis revealed: Fundamental principles underlying sensory organ diversity. Dev Biol. 2004;269:1–17. pmid:15081353
24. Pringle JWS. Proprioception In Insects. J Exp Biol. 1938;15:114–131.
25. Tuthill JC, Wilson RI. Mechanosensation and Adaptive Motor Control in Insects. Curr Biol. 2016;26:R1022–R1038. pmid:27780045
26. Prelic S, Pal Mahadevan V, Venkateswaran V, Lavista-Llanos S, Hansson BS, Wicher D. Functional Interaction Between Drosophila Olfactory Sensory Neurons and Their Support Cells. Front Cell Neurosci. 2022;15:555. pmid:35069116
27. Schmidt HR, Benton R. Molecular mechanisms of olfactory detection in insects: Beyond receptors: Insect olfactory detection mechanisms. Open Biol. 2020:10. pmid:33022193
28. Ebbs ML, Amrein H. Taste and pheromone perception in the fruit fly Drosophila melanogaster. Pflugers Arch—Eur J Physiol. 2007;454:735–747. pmid:17473934
29. Croset V, Treiber CD, Waddell S. Cellular diversity in the Drosophila midbrain revealed by single-cell transcriptomics. elife. 2018:7. pmid:29671739
30. Allen AM, Neville MC, Birtles S, Croset V, Treiber CD, Waddell S, et al. A single-cell transcriptomic atlas of the adult Drosophila ventral nerve cord. elife. 2020:9. pmid:32314735
31. Rust K, Byrnes LE, Yu KS, Park JS, Sneddon JB, Tward AD, et al. A single-cell atlas and lineage analysis of the adult Drosophila ovary. Nat Commun. 2020;11:1–17. pmid:33159074
32. Hung RJ, Hu Y, Kirchner R, Liu Y, Xu C, Comjean A, et al. A cell atlas of the adult Drosophila midgut. Proc Natl Acad Sci U S A. 2020;117:1514–1523. pmid:31915294
33. Mittnenzweig M, Mayshar Y, Cheng S, Ben-Yair R, Hadas R, Rais Y, et al. A single-embryo, single-cell time-resolved model for mouse gastrulation. Cell. 2021;184:2825–2842.e22. pmid:33932341
34. He P, Williams BA, Trout D, Marinov GK, Amrhein H, Berghella L, et al. The changing mouse embryo transcriptome at whole tissue and single-cell resolution. Nature. 2020;583:760–767. pmid:32728245
35. Seroka A, Lai SL, Doe CQ. Transcriptional profiling from whole embryos to single neuroblast lineages in Drosophila. Dev Biol. 2022;489:21–33. pmid:35660371
36. Chari T, Weissbourd B, Gehring J, Ferraioli A, Leclère L, Herl M, et al. Whole-animal multiplexed single-cell RNA-seq reveals transcriptional shifts across Clytia medusa cell types. Sci Adv. 2021;7:1683. pmid:34826233
37. Cao J, Packer JS, Ramani V, Cusanovich DA, Huynh C, Daza R, et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science (80-). 2017;357:661–667. pmid:28818938
38. Li H, Janssens J, de Waegeneer M, Kolluru SS, Davie K, Gardeux V, et al. Fly Cell Atlas: A single-nucleus transcriptomic atlas of the adult fruit fly. Science (80-). 2022;375. pmid:35239393
39. Plass M, Solana J, Alexander Wolf F, Ayoub S, Misios A, Glažar P, et al. Cell type atlas and lineage tree of a whole complex animal by single-cell transcriptomics. Science (80-). 2018;360. pmid:29674432
40. Qiu C, Cao J, Martin BK, Li T, Welsh IC, Srivatsan S, et al. Systematic reconstruction of cellular trajectories across mouse embryogenesis. Nat Genet. 2022;54:328–341. pmid:35288709
41. Fu Y, Huang X, Zhang P, van de Leemput J, Han Z. Single-cell RNA sequencing identifies novel cell types in Drosophila blood. J Genet Genomics. 2020;47:175–186. pmid:32487456
42. Basil MC, Cardenas-Diaz FL, Kathiriya JJ, Morley MP, Carl J, Brumwell AN, et al. Human distal airways contain a multipotent secretory cell that can regenerate alveoli. Nature. 2022;604:120–126. pmid:35355013
43. Zhang MJ, Pisco AO, Darmanis S, Zou J. Mouse aging cell atlas analysis reveals global and cell type-specific aging signatures. elife. 2021:10. pmid:33847263
44. Almanzar N, Antony J, Baghel AS, Bakerman I, Bansal I, Barres BA, et al. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature. 2020;583:590–595. pmid:32669714
45. Bomidi C, Robertson M, Coarfa C, Estes MK, Blutt SE. Single-cell sequencing of rotavirus-infected intestinal epithelium reveals cell-type specific epithelial repair and tuft cell infection. Proc Natl Acad Sci U S A. 2021:118. pmid:34732579
46. Xie X, Shi Q, Wu P, Zhang X, Kambara H, Su J, et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol. 2020;21:1119–1133. pmid:32719519
47. McLaughlin CN, Brbić M, Xie Q, Li T, Horns F, Kolluru SS, et al. Single-cell transcriptomes of developing and adult olfactory receptor neurons in drosophila. elife. 2021;10:1–37. pmid:33555999
48. Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS ONE. 2018:13. pmid:30586455
49. Reed CT, Murphy C, Fristrom D. The ultrastructure of the differentiating pupal leg of Drosophila melanogaster. Wilhelm Roux Arch Dev Biol. 1975;178:285–302. pmid:28304937
50. Kerber B, Monge I, Mueller M, Mitchell PJ, Cohen SM. The AP-2 transcription factor is required for joint formation and cell survival in Drosophila leg development. Development. 2001;128:1231–1238. pmid:11262225
51. Monge I, Krishnamurthy R, Sims D, Hirth F, Spengler M, Kammermeier L, et al. Drosophila transcription factor AP-2 in proboscis, leg and brain central complex development. Development. 2001;128:1239–1252. pmid:11262226
52. Hao I, Green RB, Dunaevsky O, Lengyel JA, Rauskolb C. The odd-skipped family of zinc finger genes promotes Drosophila leg segmentation. Dev Biol. 2003;263:282–295. pmid:14597202
53. Kudron MM, Victorsen A, Gevirtzman L, Hillier LW, Fisher WW, Vafeados D, et al. The ModERN Resource: Genome-Wide Binding Profiles for Hundreds of Drosophila and Caenorhabditis elegans Transcription Factors. Genetics. 2018;208:937–949. pmid:29284660
54. Rauskolb C, Irvine KD. Notch-Mediated Segmentation and Growth Control of the Drosophila Leg. Dev Biol. 1999;210:339–350. pmid:10357895
55. Mirth C, Akam M. Joint development in the Drosophila leg: Cell movements and cell populations. Dev Biol. 2002;246:391–406. pmid:12051824
56. Godt D, Couderc JL, Cramton SE, Laski FA. Pattern formation in the limbs of Drosophila: Bric a brac is expressed in both a gradient and a wave-like pattern and is required for specification and proper segmentation of the tarsus. Development. 1993;119:799–812. pmid:7910551
57. Bourbon HMG, Benetah MH, Guillou E, Mojica-Vazquez LH, Baanannou A, Bernat-Fabre S, et al. A shared ancient enhancer element differentially regulates the bric-a-brac tandem gene duplicates in the developing Drosophila leg. PLoS Genet. 2022;18:e1010083. pmid:35294439
58. Ross J, Kuzin A, Brody T, Odenwald WF. cis-regulatory analysis of the Drosophila pdm locus reveals a diversity of neural enhancers. BMC Genomics. 2015;16. pmid:26377945
59. Loker R, Mann RS. Divergent expression of paralogous genes by modification of shared enhancer activity through a promoter-proximal silencer. Curr Biol. 2022;32:3545–3555.e4. pmid:35853455
60. Kopp A. Drosophila sex combs as a model of evolutionary innovations. Evol Dev. 2011;13:504–522. pmid:23016935
61. Natori K, Tajiri R, Furukawa S, Kojima T. Progressive tarsal patterning in the Drosophila by temporally dynamic regulation of transcription factor genes. Dev Biol. 2012;361:450–462. pmid:22079694
62. Deng M, Wang Y, Zhang L, Yang Y, Huang S, Wang J, et al. Single cell transcriptomic landscapes of pattern formation, proliferation and growth in Drosophila wing imaginal discs. Development. 2019:146. pmid:31455604
63. Everetts NJ, Worley MI, Yasutomi R, Yosef N, Hariharan IK. Single-cell transcriptomics of the Drosophila wing disc reveals instructive epithelium-to-myoblast interactions. elife. 2021:10. pmid:33749594
64. Hopkins BR, Kopp A. Evolution of sexual development and sexual dimorphism in insects. Curr Opin Genet Dev. 2021;69:129–139. pmid:33848958
65. Robinett CC, Vaughan AG, Knapp J-M, Baker BS. Sex and the Single Cell. II. There Is a Time and Place for Sex. Hawley RS, editor. PLoS Biol. 2010;8: e1000365. pmid:20454565
66. Tanaka K, Barmina O, Sanders LE, Arbeitman MN, Kopp A. Evolution of sex-specific traits through changes in HOX-dependent doublesex expression. PLoS Biol. 2011;9:e1001131. pmid:21886483
67. Kocks C, Cho JH, Nehme N, Ulvila J, Pearson AM, Meister M, et al. Eater, a transmembrane protein mediating phagocytosis of bacterial pathogens in Drosophila. Cell. 2005;123:335–346. pmid:16239149
68. Evans CJ, Hartenstein V, Banerjee U. Thicker than blood: Conserved mechanisms in Drosophila and vertebrate hematopoiesis. Dev Cell. 2003;5:673–690. pmid:14602069
69. Kurucz É, Márkus R, Zsámboki J, Folkl-Medzihradszky K, Darula Z, Vilmos P, et al. Nimrod, a Putative Phagocytosis Receptor with EGF Repeats in Drosophila Plasmatocytes. Curr Biol. 2007;17:649–654. pmid:17363253
70. Tattikota SG, Cho B, Liu Y, Hu Y, Barrera V, Steinbaugh MJ, et al. A single-cell survey of Drosophila blood. elife. 2020;9:1–35. pmid:32396065
71. Rizki MT, Rizki RM. Functional significance of the crystal cells in the larva of Drosophila melanogaster. J Biophys Biochem Cytol. 1959;5:235–240. pmid:13654442
72. Hultmark D, Andó I. Hematopoietic plasticity mapped in Drosophila and other insects. elife. 2022:11. pmid:35920811
73. Grigorian M, Mandal L, Hartenstein V. Hematopoiesis at the onset of metamorphosis: Terminal differentiation and dissociation of the Drosophila lymph gland. Dev Genes Evol. 2011;221:121–131. pmid:21509534
74. del Álamo D, Terriente J, Díaz-Benjumea FJ. Spitz/EGFr signalling via the Ras/MAPK pathway mediates the induction of bract cells in Drosophila legs. Development. 2002;129:1975–1982. pmid:11934863
75. Peng Y, Han C, Axelrod JD. Planar Polarized Protrusions Break the Symmetry of EGFR Signaling during Drosophila Bract Cell Fate Induction. Dev Cell. 2012;23:507–518. pmid:22921201
76. Mou X, Duncan DM, Baehrecke EH, Duncan I. Control of target gene specificity during metamorphosis by the steroid response gene E93. Proc Natl Acad Sci U S A. 2012;109:2949–2954. pmid:22308414
77. Held LI. A high-resolution morphogenetic map of the second-leg basitarsus in Drosophila melanogaster. Wilhelm Roux Arch Dev Biol. 1979;187:129–150. pmid:28304926
78. Lawrence PA, Struhl G, Morata G. Bristle patterns and compartment boundaries in the tarsi of Drosophila. J Embryol Exp Morpholog. 1979;51:195–208. pmid:479744
79. Held LI. Bristles induce bracts via the EGFR pathway on Drosophila legs. Mech Dev. 2002;117:225–234. pmid:12204262
80. Tobler H, Rothenbühler V, Nöthiger R. A study of the differentiation of bracts in Drosophila melanogaster using two mutations, H2 and svde. Experientia. 1973;29:370–371. pmid:4196684
81. Chanana B, Graf R, Koledachkina T, Pflanz R, Vorbrüggen G. αPS2 integrin-mediated muscle attachment in Drosophila requires the ECM protein Thrombospondin. Mech Dev. 2007;124:463–475. pmid:17482800
82. Subramanian A, Wayburn B, Bunch T, Volk T. Thrombospondin-mediated adhesion is essential for the formation of the myotendinous junction in Drosophila. Development. 2007;134:1269–1278. pmid:17314133
83. Armand P, Knapp AC, Hirsch AJ, Wieschaus EF, Cole MD. A novel basic helix-loop-helix protein is expressed in muscle attachment sites of the Drosophila epidermis. Mol Cell Biol. 1994;14:4145–4154. pmid:8196652
84. Soler C, Daczewska M, Da Ponte JP, Dastugue B, Jagla K. Coordinated development of muscles and tendons of the Drosophila leg. Development. 2004;131:6041–6051. pmid:15537687
85. Gilsohn E, Volk T. Slowdown promotes muscle integrity by modulating integrin-mediated adhesion at the myotendinous junction. Development. 2010;137:785–794. pmid:20110313
86. Wayburn B, Volk T. LRT, a tendon-specific leucine-rich repeat protein, promotes muscle-tendon targeting through its interaction with Robo. Development. 2009;136:3607–3615. pmid:19793885
87. Volohonsky G, Edenfeld G, Klämbt C, Volk T. Muscle-dependent maturation of tendon cells is induced by post-transcriptional regulation of StripeA. Development. 2007;134:347–356. pmid:17166919
88. Fernandes JJ, Celniker SE, VijayRaghavan K. Development of the indirect flight muscle attachment sites in Drosophila: Role of the PS integrins and the stripe gene. Dev Biol. 1996;176:166–184. pmid:8660859
89. Sasse S, Klämbt C. Repulsive Epithelial Cues Direct Glial Migration along the Nerve. Dev Cell. 2016;39:696–707. pmid:27997826
90. Xiong WC, Okano H, Patel NH, Blendy JA, Montell C. repo encodes a glial-specific homeo domain protein required in the Drosophila nervous system. Genes Dev. 1994;8:981–994. pmid:7926782
91. Beckervordersandforth RM, Rickert C, Altenhein B, Technau GM. Subtypes of glial cells in the Drosophila embryonic ventral nerve cord as related to lineage and gene expression. Mech Dev. 2008;125:542–557. pmid:18296030
92. Yuasa Y, Okabe M, Yoshikawa S, Tabuchi K, Xiong WC, Hiromi Y, et al. Drosophila homeodomain protein REPO controls glial differentiation by cooperating with ETS and BTB transcription factors. Development. 2003;130:2419–2428. pmid:12702656
93. Yildirim K, Petri J, Kottmeier R, Klämbt C. Drosophila glia: Few cell types and many conserved functions. Glia. 2019;67:5–26. pmid:30443934
94. Halter DA, Urban J, Rickert C, Ner SS, Ito K, Travers AA, et al. The homeobox gene repo is required for the differentiation and maintenance of glia function in the embryonic nervous system of Drosophila melanogaster. Development. 1995;121:317–332. pmid:7768175
95. Lassetter AP, Corty MM, Barria R, Sheehan AE, Hill JQ, Aicher SA, et al. Glial TGFβ activity promotes neuron survival in peripheral nerves. J Cell Biol. 2023;222:2021.09.02.458753. pmid:36399182
96. Xie X, Auld VJ. Integrins are necessary for the development and maintenance of the glial layers in the Drosophila peripheral nerve. Development. 2011;138:3813–3822. pmid:21828098
97. DeSalvo MK, Hindle SJ, Rusan ZM, Orng S, Eddison M, Halliwill K, et al. The Drosophila surface glia transcriptome: Evolutionary conserved blood-brain barrier processes. Front Neurosci. 2014;8:346. pmid:25426014
98. Stork T, Bernardos R, Freeman MR. Analysis of glial cell development and function in Drosophila. Cold Spring Harb Protoc. 2012;7:1–17. pmid:22194269
99. Wheeler SR, Banerjee S, Blauth K, Rogers SL, Bhat MA, Crews ST. Neurexin IV and Wrapper interactions mediate Drosophila midline glial migration and axonal ensheathment. Development. 2009;136:1147–1157. pmid:19270173
100. Stork T, Thomas S, Rodrigues F, Silies M, Naffin E, Wenderdel S, et al. Drosophila Neurexin IV stabilizes neuron-glia interactions at the CNS midline by binding to Wrapper. Development. 2009;136:1251–1261. pmid:19261699
101. Noordermeer JN, Kopczynski CC, Fetter RD, Bland KS, Chen WY, Goodman CS. Wrapper, a novel member of the Ig superfamily, is expressed by midline glia and is required for them to ensheath commissural axons in Drosophila. Neuron. 1998;21:991–1001. pmid:9856456
102. Suzuki T, Hasegawa E, Nakai Y, Kaido M, Takayama R, Sato M. Formation of Neuronal Circuits by Interactions between Neuronal Populations Derived from Different Origins in the Drosophila Visual Center. Cell Rep. 2016;15:499–509. pmid:27068458
103. Cripps RM, Black BL, Zhao B, Lien CL, Schulz RA, Olson EN. The myogenic regulatory gene Mef2 is a direct target for transcriptional activation by Twist during Drosophila myogenesis. Genes Dev. 1998;12:422–434. pmid:9450935
104. Stork T, Engelen D, Krudewig A, Silies M, Bainton RJ, Klämbt C. Organization and function of the blood-brain barrier in Drosophila. J Neurosci. 2008;28:587–597. pmid:18199760
105. Meyer S, Schmidt I, Klämbt C. Glia ECM interactions are required to shape the Drosophila nervous system. Mech Dev. 2014;133:105–116. pmid:24859129
106. Olofsson B, Page DT. Condensation of the central nervous system in embryonic Drosophila is inhibited by blocking hemocyte migration or neural activity. Dev Biol. 2005;279:233–243. pmid:15708571
107. Li H, Watson A, Olechwier A, Anaya M, Sorooshyari SK, Harnett DP, et al. Deconstruction of the beaten Path-Sidestep interaction network provides insights into neuromuscular system development. elife. 2017:6. pmid:28829740
108. Tan L, Zhang KX, Pecot MY, Nagarkar-Jaiswal S, Lee PT, Takemura SY, et al. Ig Superfamily Ligand and Receptor Pairs Expressed in Synaptic Partners in Drosophila. Cell. 2015;163:1756–1769. pmid:26687360
109. Stork T, Sheehan A, Tasdemir-Yilmaz OE, Freeman MR. Neuron-Glia interactions through the heartless fgf receptor signaling pathway mediate morphogenesis of drosophila astrocytes. Neuron. 2014;83:388–403. pmid:25033182
110. Wu B, Li J, Chou YH, Luginbuhl D, Luo L. Fibroblast growth factor signaling instructs ensheathing glia wrapping of Drosophila olfactory glomeruli. Proc Natl Acad Sci U S A. 2017;114:7505–7512. pmid:28674010
111. Franzdóttir SR, Engelen D, Yuva-Aydemir Y, Schmidt I, Aho A, Klämbt C. Switch in FGF signalling initiates glial differentiation in the Drosophila eye. Nature. 2009;460:758–761. pmid:19597479
112. Simon F, Fichelson P, Gho M, Audibert A. Notch and Prospero repress proliferation following cyclin E overexpression in the Drosophila bristle lineage. PLoS Genet. 2009;5:e1000594. pmid:19662164
113. Simon F, Ramat A, Louvet-Vallée S, Lacoste J, Burg A, Audibert A, et al. Shaping of Drosophila neural cell lineages through coordination of cell proliferation and cell fate by the BTB-ZF transcription factor tramtrack-69. Genetics. 2019;212:773–788. pmid:31073020
114. Sanfilippo P, Smibert P, Duan H, Lai EC. Neural specificity of the RNA-binding protein elav is achieved by post-transcriptional repression in non-neural tissues. Development. 2016;143:4474–4485. pmid:27802174
115. Held LI. Sensitive periods for abnormal patterning on a leg segment in Drosophila melanogaster. Rouxs Arch Dev Biol. 1990;199:31–47. pmid:28305528
116. Rodriguez I, Hernandez R, Modolell J, Ruiz-Gomez M. Competence to develop sensory organs is temporally and spatially regulated in Drosophila epidermal primordia. EMBO J. 1990;9:3583–3592. pmid:2120046
117. Bopp D, Jamet E, Baumgartner S, Burri M, Noll M. Isolation of two tissue-specific Drosophila paired box genes, Pox meso and Pox neuro. EMBO J. 1989;8:3447–3457. pmid:2573516
118. Awasaki T, Kimura K. pox-neuro is required for development of chemosensory bristles inDrosophila. J Neurobiol. 1997;32:707–721. pmid:9183748
119. Dambly-Chaudière C, Jamet E, Burri M, Bopp D, Basler K, Hafen E, et al. The paired box gene pox neuro: A determiant of poly-innervated sense organs in Drosophila. Cell. 1992;69:159–172. pmid:1348214
120. Nottebohm E, Usui A, Therianos S, Kimura K, ichi, Dambly-Chaudiére C, Ghysen A. The gene poxn controls different steps of the formation of chemosensory organs in drosophila. Neuron. 1994;12:25–34. pmid:8292359
121. Nottebohm E, Dambly-Chaudière C, Ghysen A. Connectivity of chemosensory neurons is controlled by the gene poxn in Drosophila. Nature. 1992;359:829–832. pmid:1436059
122. Mellert DJ, Knapp JM, Manoli DS, Meissner GW, Baker BS. Midline crossing by gustatory receptor neuron axons is regulated by fruitless, doublesex and the Roundabout receptors. Development. 2010;137:323–332. pmid:20040498
123. Lu B, LaMora A, Sun Y, Welsh MJ, Ben-Shahar Y. Ppk23-dependent chemosensory functions contribute to courtship behavior in drosophila melanogaster. PLoS Genet. 2012;8:e1002587. pmid:22438833
124. Toda H, Zhao X, Dickson BJ. The Drosophila Female Aphrodisiac Pheromone Activates ppk23+ Sensory Neurons to Elicit Male Courtship Behavior. Cell Rep. 2012;1:599–607. pmid:22813735
125. Starostina E, Liu T, Vijayan V, Zheng Z, Siwicki KK, Pikielny CW. A drosophila DEG/ENaC subunit functions specifically in gustatory neurons required for male courtship behavior. J Neurosci. 2012;32:4665–4674. pmid:22457513
126. Vijayan V, Thistle R, Liu T, Starostina E, Pikielny CW. Drosophila Pheromone-Sensing Neurons Expressing the ppk25 Ion Channel Subunit Stimulate Male Courtship and Female Receptivity. PLoS Genet. 2014;10:e1004238. pmid:24675786
127. Frenkel L, Muraro NI, Beltrán González AN, Marcora MS, Bernabó G, Hermann-Luibl C, et al. Organization of Circadian Behavior Relies on Glycinergic Transmission. Cell Rep. 2017;19:72–85. pmid:28380364
128. He Z, Luo Y, Shang X, Sun JS, Carlson JR. Chemosensory sensilla of the Drosophila wing express a candidate ionotropic pheromone receptor. PLoS Biol. 2019;17:e2006619. pmid:31112532
129. Sánchez-Alcañiz JA, Silbering AF, Croset V, Zappia G, Sivasubramaniam AK, Abuin L, et al. An expression atlas of variant ionotropic glutamate receptors identifies a molecular basis of carbonation sensing. Nat Commun. 2018;9:1–14. pmid:30315166
130. Abuin L, Bargeton B, Ulbrich MH, Isacoff EY, Kellenberger S, Benton R. Functional Architecture of Olfactory Ionotropic Glutamate Receptors. Neuron. 2011;69:44–60. pmid:21220098
131. Zhang YV, Ni J, Montell C. The molecular basis for attractive salt-taste coding in drosophila. Science (80-). 2013;340:1334–1338. pmid:23766326
132. Knecht ZA, Silbering AF, Ni L, Klein M, Budelli G, Bell R, et al. Distinct combinations of variant ionotropic glutamate receptors mediate thermosensation and hygrosensation in drosophila. elife. 2016:5. pmid:27656904
133. Li H, Horns F, Wu B, Xie Q, Li J, Li T, et al. Classifying Drosophila Olfactory Projection Neuron Subtypes by Single-Cell RNA Sequencing. Cell. 2017;171:1206–1220.e22. pmid:29149607
134. Cho JY, Chak K, Andreone BJ, Wooley JR, Kolodkin AL. The extracellular matrix proteoglycan perlecan facilitates transmembrane semaphorin-mediated repulsive guidance. Genes Dev. 2012;26:2222–2235. pmid:23028146
135. Park Y, Rangel C, Reynolds MM, Caldwell MC, Johns M, Nayak M, et al. Drosophila Perlecan modulates FGF and Hedgehog signals to activate neural stem cell division. Dev Biol. 2003;253:247–257. pmid:12645928
136. Nangia V, O’Connell J, Chopra K, Qing Y, Reppert C, Chai CM, et al. Genetic reduction of tyramine β hydroxylase suppresses Tau toxicity in a Drosophila model of tauopathy. Neurosci Lett. 2021;755:135937. pmid:33910059
137. Bechstedt S, Albert JT, Kreil DP, Müller-Reichert T, Göpfert MC, Howard J. A doublecortin containing microtubule-associated protein is implicated in mechanotransduction in Drosophila sensory cilia. Nat Commun. 2010:1. pmid:20975667
138. Fichelson P, Audibert A, Simon F, Gho M. Cell cycle and cell-fate determination in Drosophila neural cell lineages. Trends Genet. 2005;21:413–420. pmid:15927300
139. Gho M, Lecourtois M, Géraud G, Posakony JW, Schweisguth F. Subcellular localization of suppressor of hairless in Drosophila sense organ cells during Notch signalling. Development. 1996;122:1673–1682. pmid:8674407
140. Miller SW, Avidor-Reiss T, Polyanovsky A, Posakony JW. Complex interplay of three transcription factors in controlling the tormogen differentiation program of Drosophila mechanoreceptors. Dev Biol. 2009;329:386–399. pmid:19232522
141. Kavaler J, Fu W, Duan H, Noll M, Posakony JW. An essential role for the Drosophila Pax2 homolog in the differentiation of adult sensory organs. Development. 1999;126:2261–2272. pmid:10207150
142. Chung YD, Zhu J, Han YG, Kernan MJ. nompA Encodes a PNS-Specific, ZP Domain Protein Required to Connect Mechanosensory Dendrites to Sensory Structures. Neuron. 2001;29:415–428. pmid:11239432
143. Kania A, Bellen HJ. Mutations in neuromusculin, a gene encoding a cell adhesion molecule, cause nervous system defects. Roux Arch Dev Biol. 1995;204:259–270. pmid:28306121
144. Kania A, Han PL, Kim YT, Bellen H. Neuromusculin, a Drosophila Gene Expressed in Peripheral Neuronal Precursors and Muscles, Encodes a Cell Adhesion Molecule. Neuron. 1993;11:673–687. pmid:8398154
145. Wei SY, Escudero LM, Yu F, Chang LH, Chen LY, Ho YH, et al. Echinoid is a component of adherens junctions that cooperates with DE-cadherin to mediate cell adhesion. Dev Cell. 2005;8:493–504. pmid:15809032
146. Grillo-Hill BK, Wolff T. Dynamic cell shapes and contacts in the developing Drosophila retina are regulated by the Ig cell adhesion protein hibris. Dev Dyn. 2009;238:2223–2234. pmid:19504462
147. Bao S, Cagan R. Preferential adhesion mediated by hibris and roughest regulates morphogenesis and patterning in the drosophila eye. Dev Cell. 2005;8:925–935. pmid:15935781
148. Ren N, He B, Stone D, Kirakodu S, Adler PN. The shavenoid gene of Drosophila encodes a novel actin cytoskeleton interacting protein that promotes wing hair morphogenesis. Genetics. 2006;172:1643–1653. pmid:16322503
149. Shapira S, Bakhrat A, Bitan A, Abdu U. The Drosophila javelin Gene Encodes a Novel Actin-Associated Protein Required for Actin Assembly in the Bristle. Mol Cell Biol. 2011;31:4582–4592. pmid:21930794
150. Mahajan-Miklos S, Cooley L. The villin-like protein encoded by the Drosophila quail gene is required for actin bundle assembly during oogenesis. Cell. 1994;78:291–301. pmid:8044841
151. Rushton E, Rohrbough J, Broadie K. Presynaptic secretion of mind-the-gap organizes the synaptic extracellular matrix-integrin interface and postsynaptic environments. Dev Dyn. 2009;238:554–571. pmid:19235718
152. Zhou P, Lou YX, Wang XG, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. pmid:32015507
153. Rahman SR, Terranova T, Tian L, Hines HM. Developmental Transcriptomics Reveals a Gene Network Driving Mimetic Color Variation in a Bumble Bee. Genome Biol Evol. 2021:13. pmid:33881508
154. Phillips AM, Smart R, Strauss R, Brembs B, Kelly LE. The Drosophila black enigma: The molecular and behavioural characterization of the black1 mutant allele. Gene. 2005;351:131–142. pmid:15878647
155. Ruiz-Losada M, Pérez-Reyes C, Estella C. Role of the Forkhead Transcription Factors Fd4 and Fd5 During Drosophila Leg Development. Front Cell Dev Biol. 2021;9:2072. pmid:34409041
156. Inbal A, Levanon D, Salzberg A. Multiple roles for u-turn/ventral veinless in the development of Drosophila PNS. Development. 2003;130:2467–2478. pmid:12702660
157. Xie Q, Brbic M, Horns F, Kolluru SS, Jones RC, Li J, et al. Temporal evolution of single-cell transcriptomes of drosophila olfactory projection neurons. elife. 2021;10:1–55. pmid:33427646
158. Özel MN, Simon F, Jafari S, Holguera I, Chen YC, Benhra N, et al. Neuronal diversity and convergence in a visual system developmental atlas. Nature. 2021;589:88–95. pmid:33149298
159. Janssens J, Aibar S, Taskiran II, Ismail JN, Gomez AE, Aughey G, et al. Decoding gene regulation in the fly brain. Nature. 2022;601:630–636. pmid:34987221
160. Davis FP, Nern A, Picard S, Reiser MB, Rubin GM, Eddy SR, et al. A genetic, genomic, and computational resource for exploring neural circuit function. elife. 2020:9. pmid:31939737
161. Magklara A, Lomvardas S. Stochastic gene expression in mammals: Lessons from olfaction. Trends Cell Biol. 2013;23:449–456. pmid:23689023
162. Everaerts C, Farine J-P, Cobb M, Ferveur J-F. Drosophila Cuticular Hydrocarbons Revisited: Mating Status Alters Cuticular Profiles. Marion-Poll F, editor. PLoS ONE. 2010;5:e9607. pmid:20231905
163. Khallaf MA, Cui R, Weißflog J, Erdogmus M, Svatoš A, Dweck HKM, et al. Large-scale characterization of sex pheromone communication systems in Drosophila. Nat Commun. 2021;12:1–14. pmid:34230464
164. Bittern J, Pogodalla N, Ohm H, Brüser L, Kottmeier R, Schirmeier S, et al. Neuron–glia interaction in the Drosophila nervous system. Dev Neurobiol. 2021;81:438–452. pmid:32096904
165. Schwabe T, Bainton RJ, Fetter RD, Heberlein U, Gaul U. GPCR signaling is required for blood-brain barrier formation in Drosophila. Cell. 2005;123:133–144. pmid:16213218
166. Mayer F, Mayer N, Chinn L, Pinsonneault RL, Kroetz D, Bainton RJ. Evolutionary conservation of vertebrate blood-brain barrier chemoprotective mechanisms in drosophila. J Neurosci. 2009;29:3538–3550. pmid:19295159
167. Buccitelli C, Selbach M. mRNAs, proteins and the emerging principles of gene expression control. Nat Rev Genet. 2020;21:630–644. pmid:32709985
168. Ittner E, Hartwig AC, Elsesser O, Wüst HM, Fröb F, Wedel M, et al. SoxD transcription factor deficiency in Schwann cells delays myelination in the developing peripheral nervous system. Sci Rep. 2021;11:1–11. pmid:34234180
169. Fogarty EA, Kitzman JO, Antonellis A. SOX10-regulated promoter use defines isoform-specific gene expression in Schwann cells. BMC Genomics. 2020;21:1–28. pmid:32770939
170. Turnescu T, Arter J, Reiprich S, Tamm ER, Waisman A, Wegner M. Sox8 and Sox10 jointly maintain myelin gene expression in oligodendrocytes. Glia. 2018;66:279–294. pmid:29023979
171. Finzsch M, Schreiner S, Kichko T, Reeh P, Tamm ER, Bösl MR, et al. Sox10 is required for Schwann cell identity and progression beyond the immature Schwann cell stage. J Cell Biol. 2010;189:701–712. pmid:20457761
172. Saur AL, Fröb F, Weider M, Wegner M. Formation of the node of Ranvier by Schwann cells is under control of transcription factor Sox10. Glia. 2021;69:1464–1477. pmid:33566433
173. Cossais F, Sock E, Hornig J, Schreiner S, Kellerer S, Bösl MR, et al. Replacement of mouse Sox10 by the Drosophila ortholog Sox100B provides evidence for co-option of SoxE proteins into vertebrate-specific gene-regulatory networks through altered expression. Dev Biol. 2010;341:267–281. pmid:20144603
174. Edwards JS, Swales LS, Bate M. The differentiation between neuroglia and connective tissue sheath in insect ganglia revisited: The neural lamella and perineurial sheath cells are absent in a mesodermless mutant of Drosophila. J Comp Neurol. 1993;333:301–308. pmid:8345109
175. Mackay TFC, Richards S, Stone EA, Barbadilla A, Ayroles JF, Zhu D, et al. The Drosophila melanogaster Genetic Reference Panel. Nature. 2012;482:173–178. pmid:22318601
176. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36:411–420. pmid:29608179
177. Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–3587.e29. pmid:34062119
178. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, et al. Comprehensive Integration of Single-Cell Data. Cell. 2019;177:1888–1902.e21. pmid:31178118
179. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33:495–502. pmid:25867923
180. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:1–15. pmid:31870423
181. McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: Doublet Detection in Single-Cell RNA Sequencing Data Using Artificial Nearest Neighbors. Cell Syst. 2019;8:329–337.e4. pmid:30954475
182. Buszczak M, Paterno S, Lighthouse D, Bachman J, Planck J, Owen S, et al. The Carnegie Protein Trap Library: A Versatile Tool for Drosophila Developmental Studies. Genetics. 2007;175:1505–1531. pmid:17194782
183. Wildonger J, Mann RS. Evidence that nervy, the Drosophila homolog of ETO/MTG8, promotes mechanosensory organ development by enhancing Notch signaling. Dev Biol. 2005;286:507–520. pmid:16168983
184. R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria; 2021. Available from: https://www.r-project.org/
185. Studio Team. RStudio: Integrated Development Environment for R. Boston, MA; 2020. Available from: http://www.rstudio.com/
186. Keil TA. Functional Morphology of Insect Mechanoreceptors. Microsc Res Tech. 1997;39:506–531. pmid:9438251
187. Keil TA. Comparative morphogenesis of sensilla: A review. Int J Insect Morphol Embryol. 1997;26:151–160.
188. Fichelson P, Gho M. The glial cell undergoes apoptosis in the microchaete lineage of Drosophila. Development. 2003;130:123–133. pmid:12441297
189. Usui T, Shima Y, Shimada Y, Hirano S, Burgess RW, Schwarz TL, et al. Flamingo, a seven-pass transmembrane cadherin, regulates planar cell polarity under the control of Frizzled. Cell. 1999;98:585–595. pmid:10490098
190. O’Connor-Giles KM, Skeath JB. Numb inhibits membrane localization of sanpodo, a four-pass transmembrane protein, to promote asymmetric divisions in Drosophila. Dev Cell. 2003;5:231–243. pmid:12919675
191. Upadhyay A, Kandachar V, Zitserman D, Tong X, Roegiers F. Sanpodo controls sensory organ precursor fate by directing notch trafficking and binding γ-secretase. J Cell Biol. 2013;201:439–448. pmid:23609534
192. Babaoglan AB, O’Connor-Giles KM, Mistry H, Schickedanz A, Wilson BA, Skeath JB. Sanpodo: A context-dependent activator and inhibitor of Notch signaling during asymmetric divisions. Development. 2009;136:4089–4098. pmid:19906847
193. Bishop SA, Klein T, Martinez Arias A, Couso JP. Composite signalling from Serrate and Delta establishes leg segments in Drosophila through Notch. Development. 1999;126:2993–3003. pmid:10357942
194. Tsuji T, Sato A, Hiratani I, Taira M, Saigo K, Kojima T. Requirements of Lim1, a Drosophila LIM-homeobox gene, for normal leg and antennal development. Development. 2000;127:4315–4323. pmid:11003832
195. Cifuentes FJ, García-Bellido A. Proximo-distal specification in the wing disc of Drosophila by the nubbin gene. Proc Natl Acad Sci U S A. 1997;94:11405–11410. pmid:9326622
196. Chu J, Si Dong PD, Panganiban G. Limb type-specific regulation of bric a brac contributes to morphological diversity. Development. 2002;129:695–704. pmid:11830570
197. Villano JL, Katz FN. four-jointed is required for intermediate growth in the proximal-distal axis in Drosophila. Development. 1995;121:2767–2777. pmid:7555705
198. Asahina K, Watanabe K, Duistermars BJ, Hoopfer E, Roberto González C, Arna Eyjólfsdóttir E, et al. Tachykinin-Expressing Neurons Control Male-Specific Aggressive Arousal in Drosophila. Cell. 2014 [cited 2022 Jul 20];156(0):221–235. pmid:24439378
199. Scott K, Brady R, Cravchik A, Morozov P, Rzhetsky A, Zuker C, et al. A chemosensory gene family encoding candidate gustatory and olfactory receptors in Drosophila. Cell. 2001;104:661–673. pmid:11257221
200. Dunipace L, Meister S, McNealy C, Amrein H. Spatially restricted expression of candidate taste receptors in the Drosophila gustatory system. Curr Biol. 2001;11:822–835. pmid:11516643
201. Robertson HM, Warr CG, Carlson JR. Molecular evolution of the insect chemoreceptor gene superfamily in Drosophila melanogaster. Proc Natl Acad Sci U S A. 2003;100:14537–14542. pmid:14608037
202. Clyne PJ, Warr CG, Carlson JR. Candidate taste receptors in Drosophila. Science (80-). 2000;287:1830–1834. pmid:10710312
203. Lilly B, Zhao B, Ranganayakulu G, Paterson BM, Schulz RA, Olson EN. Requirement of MADS domain transcription factor D-MEF2 for muscle formation in Drosophila. Science (80-). 1995;267:688–693. pmid:7839146
About the Authors:
Ben R. Hopkins
Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Software, Validation, Visualization, Writing – original draft, Writing – review & editing
* E-mail: [email protected]
Affiliation: Department of Evolution and Ecology, University of California, Davis, California, United States of America
ORCID logo https://orcid.org/0000-0002-9760-6185
Olga Barmina
Roles Investigation, Methodology, Validation
Affiliation: Department of Evolution and Ecology, University of California, Davis, California, United States of America
Artyom Kopp
Roles Conceptualization, Data curation, Funding acquisition, Methodology, Project administration, Resources, Supervision, Writing – review & editing
Affiliation: Department of Evolution and Ecology, University of California, Davis, California, United States of America
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2023 Hopkins et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
To respond to the world around them, animals rely on the input of a network of sensory organs distributed throughout the body. Distinct classes of sensory organs are specialized for the detection of specific stimuli such as strain, pressure, or taste. The features that underlie this specialization relate both to the neurons that innervate sensory organs and the accessory cells they comprise. To understand the genetic basis of this diversity of cell types, both within and between sensory organs, we performed single-cell RNA sequencing on the first tarsal segment of the male Drosophila melanogaster foreleg during pupal development. This tissue displays a wide variety of functionally and structurally distinct sensory organs, including campaniform sensilla, mechanosensory bristles, and chemosensory taste bristles, as well as the sex comb, a recently evolved male-specific structure. In this study, we characterize the cellular landscape in which the sensory organs reside, identify a novel cell type that contributes to the construction of the neural lamella, and resolve the transcriptomic differences among support cells within and between sensory organs. We identify the genes that distinguish between mechanosensory and chemosensory neurons, resolve a combinatorial transcription factor code that defines 4 distinct classes of gustatory neurons and several types of mechanosensory neurons, and match the expression of sensory receptor genes to specific neuron classes. Collectively, our work identifies core genetic features of a variety of sensory organs and provides a rich, annotated resource for studying their development and function.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer