Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Re-Classification of Drosophila melanogaster Trichoid and Intermediate Sensilla Using Fluorescence-Guided Single Sensillum Recording

  • Chun-Chieh Lin,

    Affiliation The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

  • Christopher J. Potter

    cpotter@jhmi.edu

    Affiliation The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

Abstract

Drosophila olfactory receptor neurons are found within specialized sensory hairs on antenna and maxillary palps. The linking of odorant-induced responses to olfactory neuron activities is often accomplished via Single Sensillum Recordings (SSR), in which an electrode inserted into a single sensory hair records the neuronal activities of all the neurons housed in that sensillum. The identification of the recorded sensillum requires matching the neuronal responses with known odor-response profiles. To record from specific sensilla, or to systematically screen all sensillar types, requires repetitive and semi-random SSR experiments. Here, we validate an approach in which the GAL4/UAS binary expression system is used for targeting specific sensilla for recordings. We take advantage of available OrX-Gal4 lines, in combination with recently generated strong membrane targeted GFP reporters, to guide electrophysiological recordings to GFP-labeled sensilla. We validate a full set of reagents that can be used to rapidly screen the odor-response profiles of all basiconic, intermediate, and trichoid sensilla. Fluorescence-guided SSR further revealed that two antennal trichoid sensilla types should be re-classified as intermediate sensilla. This approach provides a simple and practical addition to a proven method for investigating olfactory neurons, and can be extended by the addition of UAS-geneX effectors for gain-of-function or loss-of-function studies.

Introduction

Drosophila olfactory receptor neurons on the third antennal segments and maxillary palps are housed in specialized porous sensory hairs called sensilla. Scanning electron and light microscopy studies have revealed that these sensory hairs can be classified based on position, size, shape, and pore densities into 4 categories: basiconic, trichoid, intermediate, and coeloconic [1]. Each sensillum contains between one to four olfactory receptor neurons (ORNs). Most ORNs express only one kind of tuning Odorant Receptor (OrX) in combination with the Olfactory Co-Receptor Orco [2]. There are ~1200 ORNs in each antenna, housed in the four sensillar categories: basiconic (ab1-ab10), trichoid (at1-at4), intermediate (ai1), and coeloconic (ac1-ac4) sensilla [3]. The maxillary palps contain an order of magnitude fewer ORNs (~120) and all of the ORNs are housed in basiconic sensilla (pb1-pb3). Direct extracellular electrophysiological recordings from sensilla have proven to be a powerful technique to understand tuning affinities of individual ORNs [47]. In the single sensillum recordings (SSR) technique, a recording electrode is inserted into the base of the sensillum [4]. This allows for extracellular-field potential measurements of action potentials generated by all the ORNs within a single sensillum, and provides a quantitative method to investigate olfactory responses to stimuli.

Given the diversity of sensillar types (19 types in antenna and 3 types in maxillary palps), identification of each target sensilla for measurement of olfactory responses can be challenging and time-consuming (Fig 1). One method to circumvent this is to use the GAL4/UAS binary expression system [8, 9] to mis-express individual ORs in an easily identified basiconic sensilla (ab3) that houses a neuron lacking its endogenous receptor (the empty neuron system, genotype: Δhalo/Δhalo; Or22a-Gal4/UAS-OrX; [10]). This has proved to be an extremely powerful approach to define the odor profiles of many ORs [6, 7, 11]. However, several odorant receptors do not function in the empty neuron system (e.g, pheromone receptors, [12]), possibly because essential factors are lacking in the ab3A neuron or absent in the lymph surrounding the neuron [13, 14]. Therefore, direct measurement of neuronal activities in native sensilla remains the most reliable way to characterize the odor-activation profiles of individual ORNs. However, this can be particularly challenging for olfactory neurons that are sparse, or if odor-properties are not well defined.

thumbnail
Fig 1. Flow chart comparing traditional SSR with Fluorescence-guided SSR.

Traditional Single Sensillum Recordings (SSR) often requires screening many sensilla for odorant activities before identifying a correct target. Fluorescence-guided SSR (FgSSR) allows for GFP-labeled sensilla to be targeted directly for recordings. This increases the success rate for recording from a specific sensillum (e.g., ab4), and decreases the effort needed to record from the desired sensillum. FgSSR is also useful for targeting sparse sensilla (e.g. ab4 to ab10), sensilla with similar sizes and locations (ab6-10 with ai1-3; pb1-3), sensilla that house neurons with unknown odor-responses (e.g, ai1), or for targeting specific Odorant Receptors in unknown sensillar types. Thick “Yes/No” arrows indicate higher probabilities and thin decision arrows indicate lower probabilities.

https://doi.org/10.1371/journal.pone.0139675.g001

The identification of Drosophila odorant receptors in the sequenced genome has allowed rapid progress in characterizing the olfactory system in this model organism [15, 16]. Using the odorant receptor promoter to drive expression of GAL4 (OrX-Gal4) has allowed molecular biological approaches to match odorant receptors to olfactory neurons within defined sensilla [3, 10, 17]. However, SSR approaches have yet to take full advantage of these available molecular reagents. Here, we describe Fluorescence-guided SSR (FgSSR) in which we incorporate genetic labeling techniques with electrophysiological recordings. The FgSSR approach has many advantages over traditional SSR: 1) it increases the efficiency of screening antennal and maxillary palp sensilla for odorant responses; 2) it increases the success rate for recording from any specific sensillum; and 3) it enables recording from genetically indentified but not morphologically distinct sensilla. FgSSR is useful for targeting sparse sensilla and sensilla that house neurons with unknown odor-responses. Furthermore, by comparing the sizes and shapes of the sensilla, followed by electrophysiological recordings, we identified that intermediate and trichoid sensilla have been misclassified. Here, we present validated reagents required for FgSSR and also correct a classification of intermediate and trichoid sensilla.

Materials and Methods

Fluorescence-guided Single Sensillum Recording (FgSSR)

Sensilla of targeted ORNs were identified using 10x and 50x objectives with an optovar 1.6x attachment (Zeiss, EC Epiplan-Neofluar 10x, LC EC Epiplan-Neofluar 50x and Optovar Module 1.6x P&C ACR) on a Zeiss AxioExaminer D1 compound microscope, using a light source and eGFP filter cube (FL Filter Set 38 HE GFP shift free). Green fluorescence signals in flies were visualized from OrX-Gal4 and 10xUAS-IVS-mCD8GFP (Bloomington Stock #32186; for the OrX-Gal4 on Chr. II) or 15xUAS-IVS-mCD8GFP (Bloomington Stock #32193; for the OrX-Gal4 on Chr. III). The representative images shown in Fig 2A–2C and S1 Fig were taken on the recording rig. The suggested mounting positions of antenna are shown in Fig 3. The electrode was filled with Beadle-Ephrussi ringers solution (7.5g of NaCl+0.35g of KCl+0.279g of CaCl2-2H2O in 1L of H2O). Extracellular activity was recorded by inserting a glass electrode into the base of the sensillum of 4–8 day-old flies. Signals were amplified 100X (USB-IDAC System; Syntech, Hilversum, The Netherlands), inputted into a computer via a 16-bit analog-digital converter and analyzed off-line with AUTOSPIKE software (USB-IDAC System; Syntech). The low cutoff filter setting was 50Hz, and the high cutoff was 5kHz. Stimuli consisted of 1000 ms air pulses passed over odorant sources. The Δspikes/second was obtained by counting the spikes in a 1000ms window from 500 ms after odor stimuli were triggered, subtracting the spikes in a 1000ms window prior to stimulation. 10 standard odors for identification of sensillar types are acquired at highest purity and listed as follows: Ethyl acetate (Sigma #270989), Pentyl acetate (Sigma #109584), Ethyl butyrate (Sigma #W242705), Methyl salicylate (Sigma #76631), Hexanol (Sigma #471402), 1-octen-3-ol (Sigma #68225), E2-hexenal (Sigma #W256005), 2,3-butanedione (Sigma #B85307), Geranyl acetate (Sigma #45896), 2-heptanone (Sigma #537683), 11-cis vaccenyl acetate (50mg in 1ml ethanol, Cayman Chemical Company #0424297–6). The odors except cVA were diluted in mineral oil (Sigma #330779) at 1:100 and 30 μl was used for stimulation. Odors were delivered to the antenna as previously described [4, 6, 10]. Stimuli were delivered by placing the tip of an odor Pasteur pipette through a hole in a pipette (Denville Scientific Inc, 10ml pipette) that carried a purified continuous air stream (8.3 ml/s) directed at the antenna. A solenoid valve (Syntech) diverted delivery of a 1 s pulse of charcoal-filtered air (5 ml/s) to a Pasteur pipette containing odorant dissolved onto filter paper. Fresh odorant pipettes were used after no more than 3 odor presentations.

thumbnail
Fig 2. Expression of mCD8GFP in the olfactory neuron does not alter odor responses.

(A) ab2 sensilla were labeled using Or59b-Gal4 to drive 15xUAS-IVS-mCD8GFP expression. Antennae were visualized on the recording rig by differential interference contrast (DIC, top), and for GFP expression (middle), and the merged image is shown in the bottom row. (B) ab3 sensilla were labeled using Or22a-Gal4 to drive 15xUAS-IVS-mCD8GFP expression. (C) ab4 sensilla were labeled using Or56a-Gal4 to drive 15xUAS-mCD8GFP expression. In (A-C), arrowheads point to example cell body labeling, and arrows point to example sensillum labeling. (D) Comparing the SSR odor response profiles of wild-type (WT) and GFP-expressing neurons in ab2 and ab3 sensilla. The odor response profiles to 10 standard odorants plus mineral oil were examined for unlabeled WT and FgSSR-targeted ab2A, ab2B, ab3A, and ab3B neurons. Responses to all odorants were similar, with the exception of ab2A response to pentyl acetate, which was decreased in the FgSSR experiment (n = 4–6 for each recording). (E) The spontaneous activities of WT and GFP-labeled ab2A (Or59b), ab2B (Or85a), ab3A (Or22a), ab3B (Or85b), and at1 (Or67d) showed no significant differences (n = 9 for each recording). (F) Comparing the SSR odor response profiles of wild-type (WT) and GFP-expressing at1 Or67d+ neurons (Or67d-Gal4/15xUAS-mCD8GFP) to the pheromone ligand (cVA) at different pheromone concentrations (n = 5). Error bars indicate ± s.e.m. throughout.

https://doi.org/10.1371/journal.pone.0139675.g002

thumbnail
Fig 3. Suggested mounting positions and target zones.

Shown are images of mounted flies (top) and cartoons of prepared antenna (below). (A) A medial mounting position exposes ab1-3, ab7 and ab10 basiconic sensilla. (B) A posterior mounting position exposes ab4-6 and ab8-9 basiconic sensilla, as well as at1 and at4 trichoid sensilla types. (C) A lateral position exposes ai1-3 intermediate sensilla. (D) An anterior mounting position exposes at1, at4, ai2 and ai3 sensilla.

https://doi.org/10.1371/journal.pone.0139675.g003

Channel Rhodopsin Activation

Newly eclosed flies (age < 1 day old) were transferred to fly vials containing 0.4 mM of all-trans retinal (Sigma-Aldrich # R2500, dissolved in pure ethanol or DMSO) with fly food. The vials were kept in the dark for at least 4 days before experiments. 627nm LED light source (1-up LED Lighting Kit, PART #: ALK-1UP-EH-KIT) powered by an Arduino Uno (www.arduino.cc/en/Main/ArduinoBoardUno) was used to activate CsChrimson (Bloomington Stock # 55135 and Bloomington Stock # 55136). The light intensity was adjusted to 1.13 W/m2 by setting to 2 V supply using a custom Arduino program with a distance between the LED lens and fly antenna at 20 cm.

Differential Interference Contrast (DIC) and Confocal Imaging

The images shown in Fig 4A–4E were Z stacks of 3–4 DIC and confocal section images. Third antennal segments of 4–6 day-old flies were dissected in PBT solution (1X PBS with 0.3% Triton X-100) and fixed in 4% PFA (PBS: 70%, PBT: 10%, PFA (20%): 20%) for 15 minutes. Fixed antenna were washed with PBT for 20 minutes and mounted in PBT for imaging. DIC images and endogenous fluorescence were taken on a LSM 700 Confocal Microscope (Zeiss). Specimens were imaged fresh or no longer than 2 days after dissection since endogenous fluorescence signals would decay over time.

thumbnail
Fig 4. Simultaneous labeling and visualization of intermediate and trichoid sensilla by FgSSR.

GFP-labeling of sensilla allows for accurate classification of sensillar types. (A-E) OrX-Gal4 with 15xUAS-IVS-mCD8GFP clearly labels a specific olfactory neuron and sensilla. Middle panels show confocal DIC images of the antenna overlaid with the GFP signal, and right panels shows a higher magnification of the GFP-labelled sensilla. Arrowheads point to example cell body labeling, and arrows point to example sensillum labeling. (A) Ai1 sensilla identified by Or13a-Gal4. (B) Ai2 sensilla identified by Or23a-Gal4 and Or83c-Gal4. (C). Ai3 sensilla identified by Or19a-Gal4 and Or43a-Gal4. (D) At1 sensilla identified by Or67d-Gal4. (E) At4 sensilla identified by Or47b-Gal4, Or65a-Gal4, and Or88a-Gal4. (F) Cartoon showing how measurement of sensillar length is performed. (G) Characteristic lengths of intermediate and trichoid sensilla. p-values are as shown. t-test, n = 15 per genotype. Error bars indicate ± 2.5 s.e.m. throughout. Data points not within this range are plotted as circles. (H) FgSSR combined with 20xUAS-IVS-CsChrimson allows light-guided stimulation of olfactory neurons. This confirms at4C neurons express Or88a.

https://doi.org/10.1371/journal.pone.0139675.g004

Calculation of Sensillar Length

A target sensillum was first identified using OrX-Gal4 driven GFP fluorescence. The positions of the tip and base of the antenna were then recorded using DIC images. Briefly, the D length (Fig 3F) was calculated using the x and y positions between tip and base. Assuming tip position to be (x1, y1) and base to be (x2, y2), the distance would be [(x1-x2)2+(y1-y2)2]1/2. Z distance was decided by the number of fixed width confocal sections (thickness is 0.5 μm in this study). With D and Z, the sensillum length was calculated as [D2+Z2]1/2.

Results and Discussion

Fluorescence-guided SSR: coupling electrophysiological recordings with fluorescence-targeted olfactory neurons

Traditional single sensillum recordings (SSR) rely on the sizes, shapes, locations and neuronal odor responses (typically to a panel of up to 10 standard odors) to correctly identify target sensilla. This approach is often time-consuming and requires extensive training (Fig 1).

The introduction of binary expression systems into the Drosophila toolbox has revolutionized transgenic experimental approaches [9, 1820]. Odorant receptor-specific promoters can be used to identify olfactory neurons by driving fluorescent protein expression in an olfactory neuron of interest (e.g. OrX-Gal4, UAS-mCD8GFP). This approach was used successfully for targeting ab3 sensilla for SSR (Or22a-Gal4, 5xUAS-mCD8GFP) [10]. However, traditional reporter lines, such as 5xUAS-mCD8GFP, do not typically label sensilla with sufficient strength to allow sensillum identification [10]. As a result, the use of the GAL4/UAS system as a companion to SSR has not been widely adopted. Recent improvements to the UAS-mCD8GFP reporter, in which the number of GAL4 binding sites (upstream activation sequence, UAS) were increased to 10, 15, 20 and 40 copies, allowed for significant increases in labeling strength [21]. We reasoned that these new reporters might now allow for sufficient labeling of olfactory sensilla by existing OrX-Gal4 lines.

We acquired all available OrX-Gal4 lines from the Bloomington Stock Center, and validated which lines, when combined with 10xUAS-IVS-mCD8GFP (on Chromosome 2) or 15xUAS-IVS-mCD8GFP (on Chromosome 3), drove sufficient expression in only a single type of olfactory neuron and were still strong enough to allow identification of sensilla when viewed on a SSR compound microscope setup (Table 1; Material and Methods). This should not only significantly shorten the time span needed to identify small sensilla (e.g. ab4-ab10, pb1-3, ai1-3) but also provide a direct method to record from genetically identifiable sensilla (Fig 1). We call this method Fluorescence-guided Single Sensillum Recording (FgSSR).

Fluorescence protein expression does not interfere with olfactory activation patterns

Labeling of olfactory neurons within sensilla required high expression of membrane-targeted GFP. Since olfactory neurons require the proper functioning of membrane-bound receptors, we first questioned if membrane-targeted GFP expression in ORNs might interfere with the endogenous dynamics of ligand–receptor activation. A panel of 10 standard odorants for sensilla type identification [5, 6, 10] was applied to stimulate wild-type (WT) and GFP-labeled ab2A, ab2B, ab3A and ab3B neurons. There were no significant differences between the firing patterns of WT and GFP-labeled ORNs (Fig 2D). Furthermore, the basal firing rates of each GFP-labeled ORN were similar to those of WT (Fig 2E). Similarly, the stimulation of wild-type and GFP-labeled at1 neurons using the pheromone cis-vaccenyl acetate (cVA) demonstrated no differences (Fig 2F). Thus, using ORNs in the ab2, ab3, and at1 sensilla as a proof-of-principle, these results indicated that the high levels of membrane-bound fluorescence protein needed for sufficient labeling did not affect endogenous olfactory receptor activities.

We comprehensively examined and combined available OrX-Gal4 lines with 10xUAS or 15xUAS-IVS-mCD8GFP lines to cover all known basiconic, intermediate, and trichoid sensillar types (ab1-10, at1-2, ai1-3 and pb1-3; Table 1). For each sensillar type, at least one OrX-Gal4 line was examined by brain dissection to ensure specific and correct targeting to the reported glomerulus in the antennal lobes [3, 17]. We suggest specific OrX-Gal4 lines to use for targeting corresponding sensilla (Table 1). We also determined optimal mounting positions and the targeted regions for single sensillum recording (Fig 3 grey zones). A medial position exposed most basiconic sensilla (ab1-3, ab7 and ab10). A posterior mounting position covered basiconic (ab4-6 and ab8-9) and trichoid (at1 and at4) sensillar types. A lateral position exposed intermediate (ai1-3) sensilla. An anterior position was not frequently used because it is challenging to stabilize, but would expose at1, at4, ai2 and ai3 sensilla. Note that these positions are suggested positions to facilitate recording but were not the only positions in which the sensilla were located. For instance, ab4 sensilla were also detected in a medial mounting position but were more challenging to record in this position compared to a posterior position.

When performing SSR experiments, some sensilla are easily distinguished based on their olfactory neuron number, size, shape, and relative location on the antenna (e.g., ab1, ab2, and ab3), and do not necessarily require FgSSR (especially for ab1, which is the only large basiconic containing 4 neurons). However, small basiconic sensilla (ab4-ab10, pb1-pb3) and intermediate sensilla (ai1-ai3) are extremely difficult to distinguish on an SSR scope based on morphology alone. FgSSR allows each sensillum to be un-ambiguously identified.

The utility of FgSSR can be extended by including additional UAS-geneX effectors that might affect neuronal function. This could be used for loss-of-function studies (e.g., expression of a UAS-RNAi construct [23, 24]) or gain-of-function studies. As a proof-of-principle of the second approach, we simultaneously expressed mCD8GFP and CsChrimson in genetically specified neurons and identified the endowed light-activated responses of recorded olfactory neurons. CsChrimson is a Channel Rhodopsin modified to be activated by red-shifted light [25]. We were able to identify by GFP and verify the light-evoked neuronal activities generated in specific ORNs (Fig 4H). Or13a, which has no known odor ligand, was previously assigned into an intermediate sensillum containing only one neuron [10]. We verified this result by showing that the Or13a+ sensillum expressing CsChrimson contained only a single amplitude spike that was photo-activated by red light (genotype: Or13a-Gal4, 15xUAS-IVS-mCD8GFP; 20xUAS-IVS-CsChrimson) (Fig 4H). As further validation, we expressed CsChrimson and mCD8GFP using Or67d-Gal4 and Or88a-Gal4 and verified the olfactory neuron identities as at1 and at4C respectively. The examples also demonstrate the use of CsChrimson to verify the identity of an OrX-expressing neuron within a sensillum without the need for a specific odorant. These examples further demonstrate that this approach can be used for spike sorting by linking OrX expression to an olfactory neuron identity (e.g., Or88a expresses in the ‘C” neuron in at4).

Re-classification of trichoid sensilla

Trichoid sensilla are the largest cuticular apparatus in the third segment of antenna, ranging from 18 to 22 μm, and have the thickest sensillar wall (350–450 nm) [1]. Trichoid ORNs are generally tuned to detect pheromones [26] and reside in 4 different subtypes of trichoid sensilla—at1 (Or67d), at2 (Or23a, Or83c), at3 (Or2a, Or19a/b, Or43a) and at4 (Or47b, Or65a/b/c, Or88a) (Fig 4) [3, 17, 22]. The odorants that activate each trichoid ORN remain to be identified. So far, it has been found that Or67d is activated by the male pheromone cVA [22, 27], Or47b is activated by the pheromone methyl laurate and Or88a is activated by the pheromones methyl laurate and methyl myristate [28]. In contrast, the identified ligands for at2 and at3 neurons are not pheromones (Or83c responds to farnesol [12] and Or19a to citrus food odors [29]). This raised the question if at2 and at3 are indeed a category of trichoid sensilla.

During our validation of FgSSR, we found that some ORNs previously classified as “trichoid” [3] were not housed in what appeared to be trichoid sensilla. Instead, they were positioned in significantly shorter sensilla with thinner cuticles that are similar to intermediate sensilla [1]. With DIC combined with confocal images as the measurement of sensillar lengths (Fig 4A–4E), we found that these other sensilla were significantly shorter than trichoid sensilla, ranging from 10.38 ± 0.17μm for at2 and 8.49 ± 0.17 μm for at3 (as compared to traditional trichoid sensilla: 19.56 ± 0.31 μm for at1 and 17.7 ± 0.22 μm for at4) (Fig 4F and 4G). As the significant length differences between at1/at4 and at2/at3 likely underlied different categories of sensilla, we reclassified at2 and at3 into ai2 and ai3 respectively (Fig 5). To avoid confusion between past and future SSR studies, we did not rename at4. The 4 types of sensilla were distributed in the distolateral region on the antenna, with at1 in the proximal region, at4 at the distal region and intermixed ai2 and ai3 in between at1 and at4 (Fig 5A).

thumbnail
Fig 5. Re-classification of Drosophila melanogaster intermediate and trichoid sensilla.

(A) Schematic for the location of trichoid and intermediate sensilla on the antennae. (B) FgSSR analyses re-classified two trichoid sensilla (names bolded) as intermediate sensilla. Intermediate sensilla identification numbers are correlated to the number of housed neurons: ai1 contains one neuron, ai2 contains two neurons, and ai3 contains three neurons.

https://doi.org/10.1371/journal.pone.0139675.g005

Conclusions

The FgSSR approach described here can be used for rapid and systematic screening of odor responses in all basiconic, intermediate, and trichoid olfactory receptor neurons. Traditional SSR screens require a known odor-response profile to guide sensillar identification, and may easily miss sensilla that are rare, or whose response profiles can be confounded. FgSSR, in contrast, provides an un-biased approach, and does not require dependence on an odor-profile for identification, which can prove particularly useful for pheromone receptor neurons whose odorants are unknown or are difficult to acquire (e.g., ai1A Or13a+). In addition, since all sensilla of the same type are labeled during a single experiment, independent recordings from a sensilla type can be achieved more quickly and easily.

Our experiments have led to a re-classification of sensillar types by expanding the number of intermediate sensilla at the expense of trichoid sensilla (Fig 5; also see [12]). We suggest that there are two types of trichoid sensilla (at1 and at4), and three types of intermediate sensilla (ai1, ai2, ai3). Our classification scheme, in which the intermediate sensilla identification number also reflects the number of housed neurons, will increase the ease by which these sensilla are classified across different research groups.

Supporting Information

S1 Fig. Fluorescence-guided images of additional basiconic sensilla.

Ab5-ab10 sensilla were labeled using OrX-Gal4 to drive UAS-mCD8GFP (10xUAS or 15xUAS) expression. Antennae were visualized by differential interference contrast (DIC; top), and for GFP expression (middle) on the recording rig. The merged image is shown below.

https://doi.org/10.1371/journal.pone.0139675.s001

(TIF)

Acknowledgments

We thank O. Riabinina and D. Task for comments on the manuscript and Potter lab members for discussions on the FgSSR method.

Author Contributions

Conceived and designed the experiments: C-CL CJP. Performed the experiments: C-CL. Analyzed the data: C-CL. Contributed reagents/materials/analysis tools: C-CL. Wrote the paper: C-CL CJP.

References

  1. 1. Shanbhag SR, Müller B, Steinbrecht RA. Atlas of olfactory organs of Drosophila melanogaster: 1. Types, external organization, innervation and distribution of olfactory sensilla. International Journal of Insect Morphology and Embryology. 1999;28(4):377–97.
  2. 2. Larsson MC, Domingos AI, Jones WD, Chiappe ME, Amrein H, Vosshall LB. Or83b encodes a broadly expressed odorant receptor essential for Drosophila olfaction. Neuron. 2004;43(5):703–14. pmid:15339651
  3. 3. Couto A, Alenius M, Dickson BJ. Molecular, anatomical, and functional organization of the Drosophila olfactory system. Current Biology. 2005;15(17):1535–47. pmid:16139208
  4. 4. de Bruyne M, Clyne PJ, Carlson JR. Odor coding in a model olfactory organ: the Drosophila maxillary palp. J Neurosci. 1999;19(11):4520–32. pmid:10341252
  5. 5. de Bruyne M, Foster K, Carlson JR. Odor coding in the Drosophila antenna. Neuron. 2001;30(2):537–52. pmid:11395013
  6. 6. Hallem EA, Ho MG, Carlson JR. The molecular basis of odor coding in the Drosophila antenna. Cell. 2004;117(7):965–79. pmid:15210116
  7. 7. Hallem EA, Carlson JR. Coding of odors by a receptor repertoire. Cell. 2006;125(1):143–60. pmid:16615896
  8. 8. Fischer JA, Giniger E, Maniatis T, Ptashne M. GAL4 activates transcription in Drosophila. Nature. 1988;332(6167):853–6. pmid:3128741
  9. 9. Brand AH, Perrimon N. Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development. 1993;118(2):401–15. pmid:8223268
  10. 10. Dobritsa AA, van der Goes van Naters W, Warr CG, Steinbrecht RA, Carlson JR. Integrating the molecular and cellular basis of odor coding in the Drosophila antenna. Neuron. 2003;37(5):827–41. pmid:12628173
  11. 11. Syed Z, Ishida Y, Taylor K, Kimbrell DA, Leal WS. Pheromone reception in fruit flies expressing a moth&apos;s odorant receptor. Proc Natl Acad Sci USA. 2006;103(44):16538–43. pmid:17060610
  12. 12. Ronderos DS, Lin CC, Potter CJ, Smith DP. Farnesol-Detecting Olfactory Neurons in Drosophila. Journal of Neuroscience. 2014;34(11):3959–68. pmid:24623773
  13. 13. Xu P, Atkinson R, Jones DNM, Smith DP. Drosophila OBP LUSH is required for activity of pheromone-sensitive neurons. Neuron. 2005;45(2):193–200. pmid:15664171
  14. 14. Benton R, Vannice KS, Vosshall LB. An essential role for a CD36-related receptor in pheromone detection in Drosophila. Nature. 2007;450(7167):289–93. pmid:17943085
  15. 15. Vosshall LB, AMREIN H, Morozov PS, Rzhetsky A, Axel R. A spatial map of olfactory receptor expression in the Drosophila antenna. Cell. 1999;96(5):725–36. pmid:10089887
  16. 16. Vosshall LB, Wong AM, Axel R. An olfactory sensory map in the fly brain. Cell. 2000;102(2):147–59. pmid:10943836
  17. 17. Fishilevich E, Vosshall LB. Genetic and functional subdivision of the Drosophila antennal lobe. Current Biology. 2005;15(17):1548–53. pmid:16139209
  18. 18. Lai S-L, Lee T. Genetic mosaic with dual binary transcriptional systems in Drosophila. Nat Neurosci. 2006;9(5):703–9. pmid:16582903
  19. 19. Potter CJ, Tasic B, Russler EV, Liang L, Luo L. The Q system: a repressible binary system for transgene expression, lineage tracing, and mosaic analysis. Cell. 2010;141(3):536–48. pmid:20434990
  20. 20. Riabinina O, Luginbuhl D, Marr E, Liu S, Wu MN, Luo L, et al. Improved and expanded Q-system reagents for genetic manipulations. Nat Meth. 2015;12(3):219–22.
  21. 21. Pfeiffer BD, Ngo T-TB, Hibbard KL, Murphy C, Jenett A, Truman JW, et al. Refinement of tools for targeted gene expression in Drosophila. Genetics. 2010;186(2):735–55. pmid:20697123
  22. 22. Kurtovic A, Widmer A, Dickson BJ. A single class of olfactory neurons mediates behavioural responses to a Drosophila sex pheromone. Nature. 2007;446(7135):542–6. pmid:17392786
  23. 23. Dietzl G, Chen D, Schnorrer F, Su K-C, Barinova Y, Fellner M, et al. A genome-wide transgenic RNAi library for conditional gene inactivation in Drosophila. Nature. 2007;448(7150):151–6. pmid:17625558
  24. 24. Ni J- Q, Liu L- P, Binari R, Hardy R, Shim H-S, Cavallaro A, et al. A Drosophila resource of transgenic RNAi lines for neurogenetics. Genetics. 2009;182(4):1089–100. pmid:19487563
  25. 25. Klapoetke NC, Murata Y, Kim SS, Pulver SR, Birdsey-Benson A, Cho YK, et al. Independent optical excitation of distinct neural populations. Nat Meth. 2014:1–14.
  26. 26. van der Goes van Naters W, Carlson JR. Receptors and neurons for fly odors in Drosophila. Current Biology. 2007;17(7):606–12. pmid:17363256
  27. 27. Ejima A, Smith BPC, Lucas C, van der Goes van Naters W, Miller CJ, Carlson JR, et al. Generalization of courtship learning in Drosophila is mediated by cis-vaccenyl acetate. Current Biology. 2007;17(7):599–605. pmid:17363250
  28. 28. Dweck HKM, Ebrahim SAM, Thoma M, Mohamed AAM, Keesey IW, Trona F, et al. Pheromones mediating copulation and attraction in Drosophila. Proc Natl Acad Sci USA. 2015:201504527.
  29. 29. Dweck HKM, Ebrahim SAM, Kromann S, Bown D, Hillbur Y, Sachse S, et al. Olfactory Preference for Egg Laying on Citrus Substrates in Drosophila. Current Biology. 2013:1–9.