Introduction

Lipids play an essential role in regulating spermatogenesis across animals [14]. Studies in Drosophila have illuminated key roles for multiple lipid species in regulating sperm development [57]. For example, phosphatidylinositol and its phosphorylated derivatives participate in diverse aspects of Drosophila spermatogenesis including meiotic cytokinesis [1,8–11], somatic cell differentiation [12], germline and somatic cell polarity maintenance [1316], and germline stem cell (GSC) maintenance and proliferation [17]. Membrane lipids also influence sperm development [18, 19], whereas fatty acids play a role in processes such as meiotic cytokinesis [20] and sperm individualization [21, 22]. While these studies suggest key roles for membrane lipids and fatty acids during Drosophila spermatogenesis, some of which are conserved in mammals [2325], much less is known about how neutral lipids contribute to spermatogenesis.

Neutral lipids are a major lipid group that includes triglyceride and cholesterol ester, and reside within specialized organelles called lipid droplets (LD) [26]. LD are found in diverse cell types (e.g. adipocytes, muscle, liver, glia, neurons) [27,28,26], and play key roles in maintaining cellular lipid homeostasis. In nongonadal cell types, correct regulation of LD contributes to cellular energy production [2931], sequestration and redistribution of lipid precursors [3236], and regulation of lipid toxicity [3739]. The importance of LD to normal cellular function in nongonadal cell types is shown by the fact that dysregulation of LD causes defects in cell differentiation, survival, and energy production [26,37,40,41]. In the testis, much less is known about the regulation and function of neutral lipids and LD, and how this regulation affects sperm development.

Multiple lines of evidence suggest a potential role for neutral lipids and LD during spermatogenesis. First, genes that encode proteins associated with neutral lipid metabolism and LD are expressed in the testis across multiple species [4244]. Second, testis LD have been identified in mammals and flies under both normal physiological conditions [27,44–48] and after mitochondrial stress [49]. Third, loss of genes associated with neutral lipid metabolism and LD cause subfertility phenotypes in both flies and mammals [27,50–52]. While studies suggest that mammalian testis LD contribute to steroidogenesis [53, 54], the spatial, temporal, and cell-type specific requirements for neutral lipids and LD in the testis have not been explored in detail in any animal. It remains similarly unclear which genes are responsible for regulating neutral lipids and LD during spermatogenesis.

To address these knowledge gaps, we used Drosophila to investigate the regulation and function of neutral lipids and LD during sperm development. Our detailed analysis of spermatogenesis under normal physiological conditions revealed the presence of LD in early-stage somatic and germline cells in the testis. We identified triglyceride lipase brummer (bmm) as a regulator of testis LD, and showed that this represents a cell-autonomous role for bmm. Importantly, we found that the bmm-mediated regulation of testis LD was significant for spermatogenesis, as both whole-body and cell-autonomous loss of bmm caused defects in sperm development. Given that our lipidomic analysis revealed an excess accumulation of triglyceride in animals lacking bmm, and that genetically blocking triglyceride synthesis rescued many spermatogenic defects associated with bmm loss, our data suggests that bmm-mediated regulation of triglyceride is important for normal Drosophila sperm development. This reveals previously unrecognized roles for neutral lipids such as triglyceride in regulating spermatogenesis, and for bmm in regulating sperm development under normal physiological conditions. Together, these findings advance knowledge of the regulation and function of neutral lipids during spermatogenesis.

Results

Lipid droplets are present in early-stage somatic and germline cells

We previously reported the presence of small circular punctae (<1 μm) corresponding to LD near the apical tip of the testis [27]. We confirm these results in w1118 males using neutral lipid stain BODIPY (4,4-Difluoro-1,3,5,7,8-Pentamethyl-4-Bora-3a,4a-Diaza-s-Indacene) (Figure 1A). Importantly, we reproduced this spatial distribution of LD in two independent genetic backgrounds and at two additional ages (Figure 1B,1C). In all cases, LD were in a testis region that contains stem cells and early-stage somatic and germline cells (Figure 1A-A’, arrows), and in the hub, an organizing center and stem cell niche in the Drosophila testis (Figure 1A’’-A’’’, arrows) [55]. LD were largely absent from the testis region occupied by spermatocytes (Figure 1A and A’, arrowheads). While LD may contain multiple neutral lipid species[56], cholesterol-binding fluorescent polyene antibiotic filipin III did not detect cholesterol within testis LD (Figure S1A), suggesting triglyceride is the main neutral lipid in Drosophila testis LD.

Lipid droplets are present in early-stage somatic and germ cells.

(A) Testis lipid droplets (LD) in w1118 animals visualized with neutral lipid dye BODIPY. (A,A’) Scale bar=50 μm; (A’’,A’’’) scale bar=15 μm. Asterisk indicates hub in all images. Arrows point to LD; arrowheads point to spermatocytes in A,B. (B) Testis LD visualized with BODIPY in newly-eclosed males from two wild-type genotypes. Scale bars: main image=50 μm; inset image=10 μm. (C) Testis LD from w1118 animals at different times post-eclosion. Scale bars=50 μm. (D) Testis LD visualized with LipidTox Red in animals with somatic cell overexpression of GFP-LD (Tj-GAL4>UAS-GFP-LD). GFP-and LipidTox Red-positive punctae are somatic LD (D–D’’ arrows); LipidTox punctae without GFP indicate germline LD (D–D’’ arrowheads). Scale bars=10 μm. (E) Histogram showing the spatial distribution of somatic cell LD; error bars represent standard error of the mean (SEM). (F) Cumulative frequency distributions of somatic LD (blue line, data reproduced from E), zfh-1-positive somatic cells (zfh-1+ cells, orange line), and Eya-positive somatic cells (Eya+ cells, grey line). (G) Testis LD visualized with LipidTox Red in males with germline overexpression of GFP-LD (nos-GAL4>UAS-GFP-LD). GFP-and LipidTox Red-positive punctae indicate germline LD (arrows); LipidTox punctae without GFP indicate non-germline LD (arrowheads). Scale bars=10 μm. (H) Histogram representing the spatial distribution of LD within the germline; error bars represent SEM. (I) Histogram representing the spatial distribution of LD and GFP fluorescence (green line) (arbitrary units, a.u.) in a representative testis of a bam-GFP animal (panel J). (J) Testis LD in a bam-GFP animal; arrows point to LD and arrowheads point to spermatocytes. Scale bar=50 μm. See also Supplemental Figure 1.

Drosophila spermatogenesis requires the codevelopment and differentiation of two cell lineages, the germline and the somatic cells [57]. To identify LD in each lineage, we used the GAL4/UAS system to overexpress a transgene in which GFP is fused to the LD-targeting motif of motor protein Klarsicht [58] (UAS-GFP-LD). We targeted UAS-GFP-LD to somatic cells with Traffic jam (Tj)-GAL4 and to the germline using nanos (nos)-GAL4; LD were visualized using neutral lipid dye LipidTox. We found LD in the somatic cells of 0-day-old males (Figure 1D), and showed that the majority of somatic LD were <30 μm from the hub (Figure 1E). Because the somatic LD distribution coincided with a marker for somatic stem cells and their immediate daughter cells (Zinc finger homeodomain 1, Zfh-1) (Figure 1F; two-sample Kolmogorov-Smirnov test) [59], but not with a marker for late somatic cells (Eyes absent, Eya) [12, 60], our data suggests LD are present in early somatic cells. In the germline, GFP punctae corresponding to LD were found near the apical tip of the testis in 0-day-old males (Figure 1G,H). We found that the disappearance of germline LD coincided with peak expression of a GFP reporter that reflects the expression of Bag-of-marbles (Bam) protein in the testis (Bam-GFP) [61] (Figure 1I,1J). Because peak Bam expression signals the last round of transient amplifying mitotic cell cycle prior to the germline’s transition into the meiotic cell cycle [6264], our data suggests that germline LD, like somatic LD, are present at early stages of germline development.

brummer plays a cell-autonomous role in regulating testis lipid droplets

Adipose triglyceride lipase (ATGL) is a critical regulator of neutral lipid metabolism and LD [6574]. Loss of ATGL in many cell types triggers LD accumulation, and ATGL overexpression decreases LD number [30,67,68,71,73,73,75,76]. Given that the Drosophila ATGL homolog brummer (bmm) regulates testis LD induced by mitochondrial stress [49], we explored whether bmm regulates testis LD under normal physiological conditions. We first examined bmm expression in the testis by isolating this organ from flies in which a bmm promoter fragment drives GFP expression (bmm-GFP). Indeed, bmm-GFP accurately reproduces changes to bmm mRNA levels [77]. GFP expression was present in the germline of bmm-GFP testes, and we found germline GFP levels were higher in spermatocytes than at earlier stages of sperm development (Figure 2A,2B; one-way ANOVA with Tukey multiple comparison test). Supporting this, our analysis of a publicly available single-cell RNA sequencing data set of the male reproductive organ [78] suggested a similar trend in bmm mRNA levels between different stages of germline (Figure S2A,S2B) and somatic cell (Figure S2C,S2D) development. Importantly, germline GFP levels were negatively correlated with testis LD in bmm-GFP flies (Figure 2A,2C), suggesting regions with higher bmm expression had fewer LD.

bmm regulates testis lipid droplets in a cell-autonomous manner.

(A) Testis lipid droplets (LD) indicated by LipidTox Red in bmm-GFP animals. Arrows point to LD in all images. Arrowheads point to spermatocytes. Scale bars=50 μm. Asterisks indicate the hub in all images. (B) Quantification of nuclear GFP intensity in testes isolated from bmm-GFP animals (n=3). Germline stem cell (GSC), spermatogonia (SG), spermatocyte (SC). (C) Spatial distribution of LD (grey histogram) and GFP expression (green line) in testes from bmm-GFP animals as a function of distance from the hub (n=3). (D,E) LD near the apical region of the testis in bmmrev (D) or bmm1 (E) animals. (F) LD further away from the apical tip in bmm1 animals. (D–F) Scale bars=50 μm. (G) Histogram representing testis LD size distribution in bmmrev (grey) and bmm1 (orange). (H) Apical tip of the testes is at the left of the graph; individual dots represent a single LD and its relative position to the hub marked by an asterisk. Cumulative frequency distribution of the distance between LDs and the apical tip of the testes are drawn as solid lines. (I) Number of testis LD in bmmrev (grey) or bmm1 (orange) in FLP-FRT clones 3 days post-clone induction; dots represent measurements from a single clone. The number of cells in each cyst (CC) counted is indicated. See also Supplemental Figure 2.

To test whether bmm regulates testis LD, we compared LD in testes from 0-day-old males carrying a loss-of-function mutation in bmm (bmm1) to control male testes (bmmrev)[67]. bmm1 testes had significantly more LD across all LD sizes compared with control males (Figure 2D2G; Welch two-sample t-test with Bonferroni correction), and showed a significantly expanded LD distribution (Figure 2D2F,2H; two-sample Kolmogorov-Smirnov test). This suggests bmm normally restricts LD to the region near the apical tip of the testis, a role we confirm in both somatic and germline lineages (Figure S2E–S2H). Importantly, after inducing homozygous bmm1 or bmmrev clones in the testes using FLP-FRT system[79], we found bmm1 spermatocyte clones had significantly more LD at 3 days post-clone induction (Figure 2I; Welch two-sample t-test), a stage at which LD were absent from bmmrev clones. This indicates a previously unrecognized cell-autonomous role for bmm in regulating testis LD, a role we were unable to assess in somatic cells as we recovered no bmm1 somatic cell clones.

brummer plays a cell-autonomous role in regulating germline development

To determine the physiological significance of bmm-mediated regulation of testis LD, we investigated testis and sperm development in males without bmm function. In 0-day-old bmm1 males reared at 25°C, testis size was significantly smaller than in age-matched bmmrev controls (Figure S3A; Welch two-sample t-test), and the number of spermatid bundles was significantly lower (Figure S3B; Kruskal-Wallis rank sum test). Defects in testis size and sperm development were also observed in 14-day-old bmm1 males (Figure S3C,S3D Welch two-sample t-test). When the animals were reared at 29°C, a temperature that exacerbates spermatogenesis defects associated with changes in lipid metabolism [21], bmm1 phenotypes were more pronounced (Figure 3A-3C). This suggests loss of bmm affects testis development and spermatogenesis. Because similar phenotypes are observed in male mice without ATGL [52], and supplementing the diet of bmm1 males with medium-chain triglycerides (MCT) partially rescues the testis and spermatogenic defects we observed in flies (Figure S3E,S3F; one-way ANOVA with Tukey multiple comparison test), as it does in mice [52, 80], our data suggests flies are a good model to study how bmm/ATGL influences sperm development.

A cell-autonomous role for bmm in regulating spermatogenesis.

Testes isolated from bmmrev (A) and bmm1 (A’) animals raised at 29°C stained with phalloidin. Scale bars=100 μm. (B) Testis size in bmm1 and bmmrev animals raised at 29°C. (C) Spermatid bundle number in bmm1 and bmmrev testes from animals reared at 29°C. (D,E) Representative images of bmmrev (D) or bmm1 (E) testes stained with DAPI and anti-Vasa antibody. Arrows indicate germline stem cells (GSC). Scale bar=50 μm. The hub is marked by an asterisk in all images. (F) GSC number in bmm1 and bmmrev testes. (G) Proportion of GSCs that were either bmm1 or bmmrev clones at 3 and 14 days post-clone induction. (H) Representative images of bmmrev (H) and bmm1 (H’) testes carrying bam-GFP; data quantified in Figure S3J. Arrows indicate regions with high Bam-GFP. Scale bars=50 μm. (I) Representative images of bmmrev (I) or bmm1 (I’,I’’) testes stained with anti-Vasa antibody. Arrows indicate Vasa-positive cysts in bmm1 testis. Panel I’’ is magnified from the boxed region in I’. (I,I’) Scale bars=100 μm; (I’’) scale bar=50 μm. (J) Maximum projection of bmmrev (J) or bmm1 (J’) testes stained with anti-Boule antibody (green) and DAPI (blue). Scale bars=100 μm. Number of bmm1 and bmmrev spermatocyte clones (K) or post-meiotic clones (L) at 3 and 14 days post-clone induction. See also Supplemental Figure 3.

To explore spermatogenesis in bmm1 animals, we used germline-specific marker Vasa to visualize the germline in the testes of bmm1 and bmmrev males (Figure 3D,3E) [81]. We observed a significant increase in the number of germline stem cells (GSC) (Figure 3F; Kruskal-Wallis rank sum test) and higher variability in GSC number in bmm1 males (p=5.7×10-12 by F-test). Given that GSC number is affected by hub size and GSC proliferation [82, 83], we monitored both parameters in bmm1 and bmmrev controls. While hub size in bmm1 testes was significantly larger than in testes from bmmrev controls (Figure S3G,S3H; Welch two-sample t-test), the number of phosphohistone H3-positive GSC, which indicates proliferating GSC, was unchanged in bmm1 animals (Figure S3I; Kruskal-Wallis rank sum test). While this indicates a larger hub may partly explain bmm’s effect on GSC number, bmm also plays a cell-autonomous role in regulating GSC, as we recovered a higher proportion of bmm1 clones in the GSC pool compared with bmmrev clones at 14 days after clone induction (Figure 3G; Welch two-sample t-test).

Beyond GSC, we uncovered additional spermatogenesis defects in bmm1 testes. Peak Bam-GFP expression in testes from 0-day-old bmm1 and bmmrev males showed that GFP-positive cysts with were significantly further away from the hub in bmm1 testes (Figure 3H,S3J; Welch two-sample t-test). Indeed, 15/18 bmm1 testes contained Vasa-positive cysts with large nuclei in the distal half of the testis (Figure 3I, arrowheads), a phenotype not present in bmmrev testes (0/8) (p=0.0005 by Pearson’s Chi-square test). Because these phenotypes are also seen in testes with differentiation defects [13, 84], we recorded the stage of sperm development reached by the germline in bmm1 testes. Most bmm1 testes contained post-meiotic cells in males raised at 25°C (Figure S3K); however, germline development did not progress past the spermatocyte stage in most bmm1 testes from animals raised at 29°C (Figure S3K). Testes from bmm1 males reared at 25°C also had a smaller Boule-positive area (Figure 3J,S3L; Welch two-sample t-test), and fewer individualization complexes and waste bags (Figure S3M,S3N; Kruskal-Wallis rank sum test). Together, these data indicate loss of bmm delays germline development. Because we recovered fewer bmm1 spermatocyte and spermatid clones 14 days after clone induction (Figure 3K,3L; Kruskal-Wallis rank sum test), this effect on germline development represents a cell-autonomous role for bmm.

brummer-dependent regulation of testis triglyceride levels affects spermatogenesis

ATGL catalyzes the first and rate-limiting step of triglyceride hydrolysis [73,85,86]. Loss of this enzyme or its homologs leads to excess triglyceride accumulation [27,30,67,73,75] and shifts in multiple lipid classes [66,87–89]. To determine how loss of bmm affects spermatogenesis, we carried out mass spectrometry (MS)-based untargeted lipidomic profiling of bmm1 and bmmrev males. Hierarchical clustering of lipid species suggests that bmm1 and bmmrev males show distinct lipidomic profiles (Figure 4A). Overall, we detected 2464 and 1144 lipid features with high quantitative confidence in positive and negative ion modes, respectively. By matching experimental m/z, isotopic ratio, and tandem MS spectra to lipid libraries, we confirmed 293 unique lipid species (Supplemental table 1). We found 107 lipids had a significant change in abundance between bmm1 and bmmrev males (padj<0.05): 85 species were upregulated in bmm1 males and 22 lipid species were downregulated. Among differentially regulated species from different lipid classes, triglyceride had the largest residual above expected proportion (p=5.00×10-4 by Pearson’s Chi-squared test). This suggests triglyceride is the lipid class most affected by loss of bmm (Figure 4B,4C).

Loss of bmm disrupts triglyceride homeostasis and leads to spermatogenic defects.

(A) Hierarchical clustering of lipid species detected in bmmrev and bmm1 animals. (B) Histograms showing the proportion of species in each lipid class with different levels between bmm1 and bmmrev. Numbers on histograms indicate the number of species with differences in abundance. (C) Volcano plot showing fold change in abundance of triglycerides (green; 97 species) and non-triglyceride lipids (grey; 186 species) in our dataset. (D) Arrows indicate testis LD stained with LipidTox Red in bmmrev (D), bmm1 (D’), or mdyQX25/k03902; bmm1 (D’’) animals. (E) Whole testes isolated from bmmrev (E), bmm1 (E’), or mdyQX25/k03902;bmm1 (E’’) animals stained with anti-Vasa antibody (red) and DAPI (blue). Arrowheads indicate spermatid bundles. Scale bars=100 μm. (F) Testis size in bmmrev, bmm1, and mdyQX25/k03902;bmm1 animals. Spermatid bundles (G) and number of germline stem cells (H) in bmmrev, bmm1, and mdyQX25/k03902;bmm1 animals. (I) Testis size in animals with germline-specific mdy knockdown (nos-GAL4>mdy RNAi; bmm1) compared with controls (nos-GAL4>+; bmm1 and +>mdy RNAi; bmm1). See also Supplemental Figure 4.

In bmm1 males, most triglyceride species (55/97) were significantly higher. Because we observed a positive correlation between the fold increase in triglyceride abundance with both the number of double bonds (p=7.52×10-8 by Kendall’s rank correlation test; Figure S4A) and the number of carbons (p=2.77×10-10 by Kendall’s rank correlation test; Figure S4B), our data align well with bmm/ATGL’s known role in regulating triglyceride levels[67,68,73] and its substrate preference of long-chain polyunsaturated fatty acids[85]. While we also detected changes in species such as fatty acids, acylcarnitine, and membrane lipids (Figure S4CS4H), in line with recent Drosophila lipidomic data[90, 91], the striking accumulation of triglyceride in bmm1 males suggested that excess testis triglyceride in bmm1 males may contribute to their spermatogenic defects. To test this, we examined spermatogenesis in bmm1 males carrying loss-of-function mutations in midway (mdy). mdy is the Drosophila homolog of diacylglycerol O-acyltransferase 1 (DGAT1), and whole-body loss of mdy reduces whole-body triglyceride levels[9294]. Importantly, testes isolated from males lacking both bmm and mdy (genotype mdyQX25/k03902;bmm1) had fewer LD than testes dissected from bmm1 males (Figures 4D,S4I; one-way ANOVA with Tukey multiple comparison test).

We found that testes isolated from mdyQX25/k03902;bmm1 males were significantly larger and had more spermatid bundles than testes from bmm1 males (Figure 4E–G; one-way ANOVA with Tukey multiple comparison test). The elevated number of GSC in bmm1 male testes was similarly rescued in mdyQX25/k03902;bmm1 males (Figure 4H; one-way ANOVA with Tukey multiple comparison test). These data suggest that defective spermatogenesis in bmm1 males can be partly attributed to excess triglyceride accumulation. Notably, at least some of these defects are cell-autonomous: RNAi-mediated knockdown of mdy in the germline of bmm1 males partially rescued the defects in testis size (Figure 4I; Kruskal-Wallis rank sum test with Dunn’s multiple comparison test) and GSC variance (Figure S4J; p=4.5 x 10-5 and 8.2 x 10-3 by F-test from the GAL4-and UAS-only crosses, respectively). bmm-mediated regulation of testis triglyceride therefore plays a previously unrecognized role in regulating sperm development.

Discussion

In this study, we used Drosophila to gain insight into how the neutral lipids, a major lipid class, contribute to sperm development. We describe the distribution of LD under normal physiological conditions in the Drosophila testis, and show that LD are present at the early stages of development in both somatic and germline cells. While many factors are known to regulate LD in nongonadal cell types, we reveal a cell-autonomous role for triglyceride lipase bmm in regulating testis LD during spermatogenesis. Indeed, our data indicates loss of bmm delays germline differentiation leading to an accumulation of early-stage germ cells. These defects in germline differentiation can be partially explained by the excess accumulation of triglyceride in flies lacking bmm, as genetically blocking triglyceride synthesis rescues multiple spermatogenic defects in bmm mutants. Together, our data reveals previously unrecognized roles for LD and triglycerides during spermatogenesis, and for bmm as an important regulator of testis LD and germline development under normal physiological conditions.

One key outcome of our study was increased knowledge of LD regulation and function in the testis. Despite rapidly expanding knowledge of LD in cell types such as adipocytes or skeletal muscle, less is known about how LD influence spermatogenesis under normal physiological conditions. In mammals, testis LD contain cholesterol and play a role in promoting steroidogenesis [95, 96]. In flies, we show that LD are present in the testis, and that excess accumulation of these LD affects sperm development. In nongonadal cell types, triglycerides provide a rich source of fatty acids for cellular ATP production, lipid building blocks to support membrane homeostasis and growth, and metabolites that can act as signaling molecules [26]. Because ATP production, lipid precursors, and lipid signaling all play roles in supporting normal sperm development [97, 98], future studies will need to determine how each of these processes is affected when excess triglyceride accumulates in testis LD. This will provide critical insight into how triglyceride stored within testis LD contributes to overall cellular lipid metabolism during spermatogenesis. Because of the parallel spermatogenic defects we observed in bmm mutants and ATGL-deficient mice, we expect that these mechanisms will also operate in other species.

A more comprehensive understanding of neutral lipid metabolism during sperm development will also emerge from studies on the upstream signaling networks that regulate testis LD and triglyceride. Given that we show an important and cell-autonomous role for bmm in regulating testis LD and triglyceride, future studies will need to identify factors that regulate bmm in the testis. Based on public single-cell RNAseq data and the bmm-GFP reporter strain, our data suggest bmm mRNA levels are differentially regulated between early and later stages of sperm development. Candidates for mediating this regulation include the insulin/insulin-like growth factor signaling pathway (IIS), Target of rapamycin (TOR) pathway, and nuclear factor κB/Relish pathway (NFκB), as all of these pathways influence bmm mRNA levels in nongonadal cell types [99105]. Beyond mRNA levels, Bmm protein levels and post-translational modifications may also be differentially regulating during spermatogenesis. For example, studies show that the proteins encoded by bmm homologs in other animals are regulated by phosphorylation [106], mediated by kinases such as adenosine monophosphate-activated protein kinase (AMPK) and protein kinase A (PKA) [107109]. Importantly, many of these pathways, including IIS, TOR, AMPK, NFκB and possibly PKA influence Drosophila sperm development [110115]. Identifying the signaling networks that influence bmm regulation during sperm development will therefore lead to a deeper understanding of how testis LD and triglyceride are coordinated with physiological factors to promote normal spermatogenesis. Because pathways such as IIS and AMPK, and others, regulate sperm development in other species [116118], these insights may reveal conserved mechanisms that govern the regulation of cellular neutral lipid metabolism during sperm development.

Acknowledgements

We thank Dr. Ronald Kühnlein for bmm1 and bmmrev lines [67], Dr. Michael Welte for UAS-GFP-LD[58], and Dr. Kaeko Kamei for bmm-GFP [77]. We used stocks from the Bloomington Drosophila Stock Center (NIH P40OD018537) and Vienna Drosophila Resource Center (VDRC). We acknowledge critical resources and information provided by FlyBase [119] (supported by the National Human Genome Research Institute at the U.S. National Institutes of Health (U41 HG000739) and the British Medical Research Council (MR/N030117/1)). This work was supported by the Life Sciences Institutes Imaging Core, supported by the UBC GREx Biological Resilience Initiative. Funding for this study was provided by grants to EJR from the Canadian Institutes for Health Research (PJT-153072), Michael Smith Foundation for Health Research (16876), and the Canadian Foundation for Innovation (JELF-34879). GT was supported by a grant from the Natural Sciences and Engineering Research Council (NSERC; 2018-04648), TH/HY/CW were supported by NSERC (2020-04895), MA was supported by the Jacob’s foundation. We would like to acknowledge that our research takes place on the traditional, ancestral, and unceded territory of the Musqueam people; a privilege for which we are grateful.

Author contributions

Conceptualization, C.C. and E.J.R.; Methodology, C.C. and Y.Y.P.; Software, C.C.; Investigation, C.C., H.Y., and Y.Y.P.; Lipidomics, M.A., H.Y., C.W., T.H.; Writing – Original Draft, C.C. and E.J.R.; Writing – Review and Editing, C.C., E.J.R., and Y.Y.P.; Supervision, E.J.R., G.T., and T.H.; Project administration, E.J.R.; Funding Acquisition, E.J.R., G.T., and T.H.

Declaration of interests

The authors declare no competing interests.

Materials and methods

Materials and Resource availability.

Drosophila strains and their source are listed in a Key Resources table. Further information and requests for resources and reagents should be directed to, and will be fulfilled by, lead contact Dr. Elizabeth J. Rideout (elizabeth.rideout@ubc.ca).

Data and Code availability.

All raw data and results of statistical tests reported in this paper are located in Supplementary files 1-4. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Fly husbandry.

Fly stocks were maintained at room temperature in 12:12 hour light:dark cycle. Unless otherwise indicated, all flies were raised at 25°C with a density of 50 larvae per 10 mL fly media. Because this project examines sperm development, we used male flies in all experiments. Fly media contained 20.5 g sucrose (SU10, Snow Cap), 70.9 g Dextrose (SUG8, Snow Cap), 48.5 g cornmeal (AO18006, Snow Cap), 30.3 g baker’s yeast (NB10, Snow Cap), 4.55 g agar (DR-820-25F, SciMart), 0.5 g calcium chloride dihydrate (CCL302.1, BioShop Canada), 0.5 g magnesium sulfate heptahydrate (MAG511.1, BioShop Canada), 4.9 mL propionic acids (P1386, Sigma-Aldrich), and 488 μL phosphoric acid (P5811, Sigma-Aldrich) per 1L of media. For diets with medium-or long-chain triglyceride, 4 g of coconut oil (medium chain triglyceride) or olive oil (long chain triglyceride) was added per 100 mL of media described above prior to cooling. Males were collected and dissected within 24 hours of eclosion unless otherwise indicated. Fixations were performed at room temperature with 4% paraformaldehyde (CA11021-168, VWR) in PBS for 20 minutes on a rotating platform followed by washing in PBS twice before staining. Fly strains used in our study are listed in a Key Resources table.

Testis cell stage classification and measurements.

Cells at an early stage of development (stem cells and early-stage somatic and germline cells) were located in the apical region of the testis, and were identified by their small and dense nuclei[120]. GSC were defined as Vasa-positive cells in direct contact with the hub; proliferating GSC were identified as Vasa-positive cells in direct contact with the hub that were also phospho-H3 positive. Cells in the testis region occupied by primary spermatocytes were identified by their large cell size and decondensed chromosome staining occupying three nuclear domains[120]. Spermatid bundles were identified by their condensed and needle-shaped nuclei, which roughly corresponds to nuclei with protamine-based chromatin[121]. Testis size was measured by quantifying the length of a line drawn down the middle of a testis image; starting from the apical tip of the testis and ending where the testis meets the seminal vesicle.

FLP-FRT clone induction.

Adult males were collected at 3-5 days post-eclosion and heat-shocked three times at 37°C with a 10 min rest period at room temperature between heat shocks. After heat-shock, the flies were incubated at room temperature until dissection.

Immunohistochemistry.

Fixed samples were rinsed three times with blocking solution containing 0.2% bovine serum albumin (A4503, Sigma-Aldrich), 0.3% Triton-X in PBS, then blocked for 1 hr on a rotating platform at room temperature. During the incubation, the blocking solution was changed every 15 minutes. After blocking, the sample were resuspended in blocking solution with the appropriate concentration of primary antibody (see Key Resources table), and incubated overnight at 4°C. Samples were rinsed three times with blocking solution after removing primary antibody, and blocked for one hour on a rotating platform in blocking solution. Secondary antibody was applied in blocking solution and left on the rotating platform at room temperature for 40 min. The sample was rinsed with blocking solution three more times, and washed four times for 15 min per wash in blocking solution. Testis samples were resuspended in Vectashield mounting media with DAPI (H-1200-10, Vector Laboratory) or SlowFade Diamond mounting media (S36972, Thermo Fisher Scientific) prior to mounting.

Lipid droplet staining.

Fixed testes were briefly permeabilized with 0.1% Triton-X in PBS for 5 min prior to applying phalloidin. For BODIPY staining, samples were suspended in PBS containing 10 μg/mL DAPI (2879083-5mg, PeproTech), 1:500 BODIPY 495/503 (Thermo Fisher Scientific D3922), and 1:1000 phalloidin iFluor647 (ab176759, Abcam) or 1:40 phalloidin TexasRed (T7471, Thermo Fisher Scientific). For staining with LipidTox Red, samples were suspended in PBS containing 10 μg/mL DAPI (2879083-5mg, PeproTech), 1:200 LipidTox Red (H34476, Thermo Fisher Scientific), and 1:1000 phalloidin iFluor647 (ab176759, Abcam). For staining free sterols, samples were prepared as for BODIPY staining with 50 μg/mL filipin in place of BODIPY for 30 min. Samples were incubated on a rotating platform for 40 minutes at room temperature. After incubation, samples were washed twice with PBS, then resuspended in SlowFade Diamond mounting media (Thermo Fisher Scientific S36972) prior to mounting.

Image acquisition and processing.

All images were acquired on a Leica SP5 confocal microscope system with 20X or 40X objectives and quantified with Fiji image analysis software[122].

Drosophila lipidomics.

Drosophila extracts were prepared following the previously reported protocol[123]. Briefly, 10 Drosophila males (∼10 mg) were weighed, 300 µL of ice-cold methanol/water mixture (9:1, v:v) was added to these males, and the samples were homogenized with glass beads using a bead beater (mini-beadbeater-16, BioSpec, Bartlesville, Ok, USA). Sample weight was used for sample normalization. Fly lysate was kept at-20°C for 4 hours for protein precipitation. Then, 900 µL of methyl tert-butyl ether was added and the solution was shaken for 5 min to extract lipids. To induce phase separation 285 µL of water was added, followed by centrifugation. The upper layer was separated, dried, and reconstituted in isopropanol/acetonitrile (1:1, v:v) for liquid chromatography-mass spectrometry (LC-MS) analysis. The volume of reconstitution solution was proportional to sample weight for normalization. Quality control (QC) samples were prepared by pooling 20 μL aliquot from each sample. The method blank sample was prepared using an identical workflow but without adding Drosophila.

Drosophila extracts were analyzed on an UHR-QqTOF (Ultra-High Resolution Qq-Time-Of-Flight) mass spectrometry Impact II (Bruker Daltonics, Bremen, Germany) interfaced with an Agilent 1290 Infinity II LC Systems (Agilent Technologies, Santa Clara, CA, USA). LC separation was performed using a Waters reversed-phase (RP) UPLC Acquity BEH C18 Column (1.7 µm, 1.0 mm ×100 mm, 130 Å) (Milford, MA, USA) maintained at 30°C. For positive ion mode, the mobile phase A was 60% acetonitrile in water and the mobile phase B was 90% isopropanol in acetonitrile, both containing 5 mM ammonium formate (pH = 4.8, adjusted by formic acid). For negative ion mode, the mobile phase A was 60% acetonitrile in water and the mobile phase B was 90% isopropanol in acetonitrile, both containing 5 mM NH4FA (pH = 9.8, adjusted by ammonium hydroxide). The LC gradient for positive and negative ion modes was set as follows: 0 min, 5% B; 8 min, 40% B; 14 min, 70% B; 20 min, 95% B; 23 min, 95% B; 24 min, 5% B; 33 min, 5% B. The flow rate was 0.1 mL/min. The injection volume was optimized to 2 µL in positive mode and 5 µL in negative mode using QC sample. The ESI source conditions were set as follows: dry gas temperature, 220 °C; dry gas flow, 7 L/min; nebulizer gas pressure, 1.6 bar; capillary voltage, 4500 V for positive mode and 3000 V for negative mode. The MS1 analysis was conducted using following parameters: mass range, 70-1000 m/z; spectrum type: centroid, calculated using maximum intensity; absolute intensity threshold: 250. Data-dependent MS/MS analysis parameters: collision energy: 16-30 eV; cycle time, 3 s; spectra rate: 4 Hz when intensity < 104 and 12 Hz when intensity > 105, linearly increased from 104 to 105. External calibration was applied using sodium formate to ensure the m/z accuracy before sample analysis.

The raw LC-MS data were processed using MS-DIAL (ver. 4.38)[124]. The detailed MS-DIAL parameters are: MS1 tolerance, 0.01 Da; MS/MS tolerance, 0.05; mass slice width, 0.05 Da; smoothing method, linear weighted moving average; smoothing level, 3 scans; minimum peak width, 5 scans. Lipid features with high quantitative confidence were selected by the following criteria: retention time was within the gradient elution time (< 23 min); average intensity in QC samples is larger than 5-fold of the intensity in method blank sample. Lipid identification was performed by matching experimental precursor m/z, isotopic ratio and MS/MS spectrum against the LipidBlast libraries embedded in MS-DIAL. To improve the quantification accuracy, the measured MS signal intensities were corrected using serial diluted QC samples following the reported workflow[125].

Quantification and statistical analysis.

All microscopy images were quantified using Fiji software[122]. For lipid droplet counts, a single optical slice through the middle of the testis containing the hub was used with the exception of FLP-FRT experiment where all lipid droplets within a GFP-negative cyst were counted (Figure 2I). All statistical analyses were done using R (obtained from https://cran.r-project.org). With exception of data concerning spatial distribution, and lipidomic data, Shapiro-Wilk test (via shapiro.test in base R) was used to assess normality of distribution prior to testing for significance. Kruskal-Wallis rank sum test (from the R package coin) and Dunn’s test (from the R package dunn.test) were used in place of Welch two-sample t-test and Tukey’s multiple comparison test when the assumption of normality was not met. For testing differences in variance between two populations, F-test (via var.test in base R) was used. For testing differences in spatial distribution, two-sample Kolmogorov-Smirnov test (via ks.test in base R) was used. All p-values are indicated in figures; extremely small p-values are listed as p<2.2 x 10-16.

Resource table

Supplemental table and files

Supplemental table 1 – Table showing identified lipid species from untargeted lipidomic analysis.

Supplementary file 1 – Raw data and statistical outputs from Figure 1.

Supplementary file 2 – Raw data and statistical outputs from Figure 2 and Supplemental figure 2.

Supplementary file 3 – Raw data and statistical outputs from Figure 3 and Supplemental figure 3.

Supplementary file 4 – Raw data and statistical outputs from Figure 4 and Supplemental figure 4.

Supplemental figure legends

Cholesterol is absent from testis lipid droplets.

(A) Testes stained with BODIPY (A) to detect neutral lipids and Filipin III (A’) to detect free cholesterol. Scale bars=50 μm.

bmm mRNA levels during spermatogenesis in germline and somatic lineages.

(A) Pseudotime trajectory of germline (black line) based on single-cell RNA sequencing data62. Individual cells are labeled according to the annotation within the data set. (B) Rolling average of normalized bmm transcript counts in the germline along the trajectory shown in panel A are plotted as a black line on the upper panel. Composition of cell types mapped on to the trajectory at each time point is shown at the lower half of panel B. (C) Pseudotime trajectory of the somatic cells (black line) based on publicly available single-cell RNA sequencing data62. Individual cells are labeled according to the annotation within the data set. (D) Rolling average of normalized bmm transcript counts in somatic cells plotted as a black line along the trajectory shown in C (upper panel). Composition of cell types mapped on to the trajectory at each time point (lower panel). (E-H) Representative images of bmmrev (E and F) and bmm1 (G and H) testes with somatic over-expression of GFP-LD (Tj-GAL4>UAS-GFP-LD). Panel F and H contain magnified images of the area indicated by the boxes in panel E and G, respectively. In bmmrev testes, LD were restricted to a region near the apical tip (E) of the testis in both somatic (F–F’’’ arrows) and germline cells (F–F’’’ arrowheads). In bmm1 testes, LD were present in both somatic (G–H arrows) and germline cells (G–H arrowheads), near the apical tip of the testis in a region corresponding to early-stage germ cells and in the region corresponding to spermatocytes. (E,G) Scale bars=50 μm; (F,H) scale bars=20 μm.

Additional characterization of testis development and spermatogenesis in animals lacking bmm.

(A) Testis size was smaller in bmm1 mutant animals compared with bmmrev controls at <24 hr post-eclosion when raised at 25°C (A; Welch two-sample t-test). (B) The number of spermatid bundles was significantly lower in bmm1 mutant animals compared with bmmrev controls at <24 hr post-eclosion when raised at 25°C (Kruskal-Wallis rank sum test). (C) Testis size was significantly smaller in bmm1 mutant males compared with bmmrev control males at 14-days post-eclosion (Welch two-sample t-test). (D) While the median number of spermatid bundles was not significantly different between bmm1 mutant males and bmmrev control males at 14 days post-eclosion (Welch two-sample t-test), 8/27 bmm1 testis had no spermatid bundles, a phenotype absent in age-matched bmmrev males (0/22) (p=0.0163, Pearson’s Chi-squared test), suggesting a subtle defect is present. (E) Food supplemented with 4% medium chain triglyceride (MCT), but not long chain triglyceride (LCT), significantly increased testis length in bmm1 animals but had no effect on this phenotype in bmmrev control animals (one-way ANOVA with Tukey multiple comparison test). (F) Food supplemented with 4% medium chain triglyceride significantly increased the number of spermatid bundles in bmm1 testes but had no effect on this phenotype in bmmrev control animals (one-way ANOVA with Tukey multiple comparison test). (G) Representative images of bmmrev (G–G’) or bmm1 (G’’– G’’’) testes stained for Fas3 (G and G’’) and Vas (G’ and G’’’). Scale bars=25 μm. (H) Quantification of hub area in bmmrev or bmm1 testes showed a significantly larger hub size in bmm1 testes (Welch two-sample t-test). (I) The number of germline stem cells (GSC) undergoing mitosis (phospho-histone H3+ GSC/total GSC) was not significantly different between bmm1 and bmmrev testes (Kruskal-Wallis rank sum test). (J) The distance between the hub and the first Bam-GFP positive cyst (Figure 3H) was significantly higher in bmm1 testes than in bmmrev testes (Welch two-sample t-test). (K) All bmmrev testes and most bmm1 testes contained spermatids when raised at 25°C; however, the most advanced stage of spermatogenesis observed in the majority of bmm1 testes isolated from animals reared at 29°C was the spermatocyte stage. (L) Testes isolated from bmm1 animals showed a significantly smaller Boule-positive area than control testes (Welch two-sample t-test). (M) Testes isolated from bmm1 animals contain fewer individualization complexes than bmmrev control testes (Kruskal-Wallis rank sum test). (N) Fewer waste bags were present in testes isolated from bmm1 animals compared with bmmrev control testes (Kruskal-Wallis rank sum test).

Lipidomic analysis of animals lacking bmm.

(A) Higher fold-changes of triglycerides in bmm1 animals were associated with less saturation in the acyl-groups (Kendall’s rank correlation test). (B) Higher fold-changes of triglycerides in bmm1 animals were associated with higher number of carbons in the acyl-groups (Kendall’s rank correlation test). Each dot represents a single triglyceride species for panel B and C. (C) Volcano plot of identified lipids; monoglycerides shown in blue and diglycerides shown in orange. Many monoglycerides and diglycerides show increase in fold-change in bmm1 males. (D) The number of carbon and the degree of saturation of monoglycerides (MAG) and diglycerides (DAG) with significant changes in abundance between bmm1 and bmmrev males. (E) Volcano plot of identified lipids; fatty acids shown in magenta and acyl-carnitine shown in green. Many fatty acids show an increase in fold-change while many acyl-carnitines show a decrease in fold-change in bmm1 males. (F) The number of carbon and the degree of saturation of fatty acids (FA) and acyl-carnitines (ACar) with significant changes in abundance between bmm1 and bmmrev males. (G) Volcano plot of identified lipids; membrane lipids shown in yellow. (H) The number of carbon and the degree of saturation of membrane lipids with significant changes in abundance between bmm1 and bmmrev males. For panel G and H, PC: phosphatidylcholine; PE: phosphatidylethanolamine; PI: phosphatidylinositol; LPC: lysophosphatidylcholine; LPE: lysophosphatidylethanolamine; SM: sphingomyelin; PG: phosphatidylglycerol. (I) Loss of mdy function rescued the elevated number of LD in bmm1 testes to control levels (one-way ANOVA with Tukey multiple comparison test). (J) Germline-specific loss of mdy in bmm1 animals did not reduce GSC numbers, but the variance in GSC number was significantly rescued (nos-GAL4>+; bmm1 vs nos-GAL4>mdy RNAi; bmm1: p=4.5 × 10-5; +>mdy RNAi; bmm1 vs nos-GAL4>mdy RNAi; bmm1: p=0.0082 by F-test).