Skip to main content

Drosophila melanogaster positive transcriptional elongation factors regulate metabolic and sex-biased expression in adults

Abstract

Background

Transcriptional elongation is a generic function, but is also regulated to allow rapid transcription responses. Following relatively long initiation and promoter clearance, RNA polymerase II can pause and then rapidly elongate following recruitment of positive elongation factors. Multiple elongation complexes exist, but the role of specific components in adult Drosophila is underexplored.

Results

We conducted RNA-seq experiments to analyze the effect of RNAi knockdown of Suppressor of Triplolethal and lilliputian. We similarly analyzed the effect of expressing a dominant negative Cyclin-dependent kinase 9 allele. We observed that almost half of the genes expressed in adults showed reduced expression, supporting a broad role for the three tested genes in steady-state transcript abundance. Expression profiles following lilliputian and Suppressor of Triplolethal RNAi were nearly identical raising the possibility that they are obligatory co-factors. Genes showing reduced expression due to these RNAi treatments were short and enriched for genes encoding metabolic or enzymatic functions. The dominant-negative Cyclin-dependent kinase 9 profiles showed both overlapping and specific differential expression, suggesting involvement in multiple complexes. We also observed hundreds of genes with sex-biased differential expression following treatment.

Conclusion

Transcriptional profiles suggest that Lilliputian and Suppressor of Triplolethal are obligatory cofactors in the adult and that they can also function with Cyclin-dependent kinase 9 at a subset of loci. Our results suggest that transcriptional elongation control is especially important for rapidly expressed genes to support digestion and metabolism, many of which have sex-biased function.

Background

Transcription by eukaryotic RNA polymerase II complex (RNAPII), composed of ten or more subunits [1, 2], involves multiple steps including initiation, promoter clearance, elongation, and termination. Each step requires different activities. The initiation process involves RNAPII recognition of core promoter elements at the transcription start site (TSS) [3]. Bound RNAPII opens a bubble in the duplex DNA and begins transcribing short RNAs by a scrunching mechanism [4]. While the first steps of initiation and promoter clearance are slow and highly regulated, elongation is rapid [5, 6]. This makes elongation an ideal mechanism for rapidly responding to changing conditions. A stalled or paused RNAPII has completed the slow steps such that de-pausing allows for rapid completion of transcription.

Great progress has been made on the biochemistry of elongation, although the exact composition of complexes is unclear due to biological context, biochemical complexity, and/or methodologies used in various studies. For the majority of genes, after transcription of about 20 to 50 nucleotides, RNAPII pauses when 5,6-dichloro-1-β-D-ribofuranosylbenzimidazole (DRB) sensitivity inducing factor (DSIF, a heterodimer of Suppressor of Ty 4 and 5 (Spt4 and Spt5)) and negative elongation factor (NELF) protein complexes bind to the RNAPII and transcript [7,8,9], to form a DSIF-NELF complex promoting the pausing of RNAPII [10,11,12]. Paused RNAPII is released by the positive transcription elongation factor b complex (P-TEFb), which phosphorylates the DSIF and NELF complexes as well as the carboxy-terminal domain of RNAPII (CTD) [13,14,15,16,17]. The catalytic and regulatory components of P-TEFb are Cyclin-dependent kinase 9 (Cdk9) and Cyclin T respectively [18,19,20]. In addition to P-TEFb, Cdk9 can also form a complex with Cyclin K [21]. Recruitment of P-TEFb kinase dissociates NELF, transforming DSIF into a positive elongation factor, triggering elongation [10].

P-TEFb is often part of larger complexes (Table 1). For example, the super elongation complex (SEC), is a key regulator required for transcriptional elongation checkpoint control and rapid transcriptional induction [22,23,24]. In addition to P-TEFb, SEC contains 11-19 Lysine-rich Leukemia (ELL), encoded by Suppressor of Triplolethal (Su(Tpl)), and ALL1-Fused Gene From Chromosome 4(AF4)/Fragile mental retardation 2 (FMR2) family member 4 (AFF4), encoded by lilliputian (lilli) [23, 25, 26]. Another transcriptional elongation complex is the Dot1 complex (DotCom) [27], which contains the histone H3 Lys 79 (H3K79) methyltransferase DOT1L, encoded by grappa (gpp) [28]. H3K79 methylation by DOT1 may regulate transcription of genes by changing chromatin structure and accessibility of transcription factors [29], and has been implicated in many basic functions such as cell cycle regulation, chromosomal stability, and the DNA damage response [30]. There is also evidence that lilli and Su(Tpl) are essential for the H3K79 methylation function of DotCom during transcriptional activation, and evidence of large complexes including components of DOT1, P-TEFb, and SEC, such as the Elongation Assisting Protein complexes (EAP) and complexes of Mixed Lineage Leukemia 1 proteins (MLL) [23, 27, 31, 32]. Additional complexity includes the Little Elongation Complex (LEC), which contains Su(Tpl) but lacks Lilli and Cdk9 [33], and AF4 family/ENL family/P-TEFb complex (AEP), which contains Lilli and Cdk9 but lacks Su(Tpl) [34]. These studies indicate that there are many possible elongation complexes [25, 31]. We have explored the roles of these complexes by examining the RNA expression profiles when some of these key components are altered in Drosophila. Specifically, knockdown of lilli or Su(Tpl) should disrupt SEC, EAP, and MLL. LEC should be altered by knockdown of Su(Tpl) but not lilli, and AEP should be altered by knockdown of lilli but not Su(Tpl). The dominant-negative form of Cdk9 should disrupt P-TEFb, SEC, and AEP, but not MLL.

Table 1 Components of different elongation related complexes or protein group

The regulatory advantage of RNAPII pausing and elongation control is that rapid responses can be achieved if earlier rate-limiting steps occur, leaving only the more rapid elongation step for completion of transcription [5, 35,36,37]. Such a rapid transcriptional response is critical when organisms experience rapid environmental changes. For example, RNAPII pausing has been extensively studied in the context of the rapid transcription of Drosophila heat shock genes [38,39,40]. The rapid response is essential because failure to adjust to rapid temperature change results in catastrophic denaturation or mis-folding of proteins [41]. Increased levels of protein chaperones encoded by the heat shock loci ameliorate this stress. The presence of DSIF and NELF results in RNAPII pausing near the heat shock gene promoters [39], but following a heat shock, P-TEFb quickly redistributes to the heat shock genes, which allows RNAPII to elongate resulting in rapid production of transcripts [42, 43]. Other types of stress requiring a quick response in Drosophila, such as arbovirus infection, are also regulated at the level of elongation [44]. Regulation of transcription elongation also plays a role during development. An example is the pausing of RNAPII downstream of the sloppy-paired-1 promoter by NELF during the rapid blastoderm-stage of development [45]. It is likely that there are other genes responding rapidly due to normal physiology. Our expression profiles might help identify them.

Elongation control also plays a role in sexual development in Drosophila. Sex is determined by counting X chromosomes [46]. In wild-type flies, XX individuals are female and XY individuals are male. The genic imbalance created by X chromosome monosomy in males is compensated by increasing X chromosome gene expression [47]. At the regulatory level, the XX karyotype results in the activation of Sex-lethal (Sxl), which controls a major branch in the sex determination network [48]. Sxl regulates the splicing of transformer (tra) pre-mRNA, which ultimately is responsible for nearly all sexually dimorphic aspects of development, physiology, and behavior [49,50,51]. One of the most important genes downstream of tra is doublesex (dsx), which encodes sex-specific isoforms of a doublesex-mab3-related-transcription factor (DMRT). DMRTs are used to regulate sexual dimorphism in most metazoans [52]. The lilli and Su(Tpl) loci are likely to be direct targets of Dsx, and while the Cdk9 locus was not among the most obvious Dsx target genes, it has a strong and conserved Dsx binding site [51, 53]. Knocking down lilli or Su(Tpl) in dsx expressing cells results in dramatic sex- and context-specific phenotypes. For example, in males, the formation of specific first leg bristles known as sex combs, shows female pigmentation and narrower female-like morphology, but male distal/proximal orientation. In females, the first legs are normal, but bristles on the genitalia are missing. In males, the genitalia are normal. Thus, these genes function in female-specific development in one tissue and male-specific development in another [51]. If regulators of elongation play a post-developmental role in other aspects of sexual dimorphism, such as physiology, then we would expect to see sex-specific effects of disrupting the function of those complexes in adult flies.

Results

Knockdown and overexpression efficiency

We were interested in measuring the effect of lilli, Su(Tpl), and Cdk9 on gene expression in adults to probe for roles of elongation. Because these genes are required for organismal viability during development [54, 55], we bypassed the developmental defects by using ubiquitously expressed Gal4 [56] along with ubiquitously expressed temperature sensitive Gal80 repressor (Gal80 ts) [57] and Gal4 responsive constructs expressing RNAi against lilli, and Su(Tpl) [58] (Fig. 1a). We also used a dominant negative Cdk9 construct (Cdk9 DN) made by substituting an asparagine for an aspartic acid codon at position 199 (D199N) in the active site of the kinase. The same substitution has been used in other studies, where it has been shown to block P-TEFb CTD kinase activity [59, 60]. Cdk9 DN under UAS control was introduced into flies using P-element mediated transformation. Expressing Cdk9 DN during development resulted in lethality (DB and DP, unpublished data). Flies bearing the Cdk9 DN transgene were viable to adulthood at 20 °C in the presence of both Gal4 and Gal80 ts.

Fig. 1
figure 1

Experimental design and efficiency of RNAi or Cdk9 DN expression. a Experimental design. The cartoons (top) illustrate the sham genotypes where Gal80ts (red) and Gal4 (green) are produced in the absence of a responding UAS transgene (left), or when the UAS transgenes are present at the uninducing (middle) or inducing (right) temperatures. We used homozygous P{tubP-GAL4} LL7, P{tubP-GAL80 ts}7 virgin females for all crosses. Sham flies were produced by crossing to lacZ (Sham1, yellow fill) or w 1118 (Sham2, gray fill) males. The schemata (bottom) shows how the shams were used as references for all induction timepoints for both females and males in results presented in subsequent figures. See Methods for further information. Transgenes are shown as bars, with regulatory sequences (open) from tubulin (tub) or the Gal4 upstream activation sequence (UAS), and encoding sequences (filled) from the Gal80 ts (red), Gal4 (green), shRNAi, and Cdk9 DN (black) labeled. Gal80ts (red hexagons), Gal4 (green ovals) proteins are shown. Active (bent black arrows) and repressed (red X) transcription are indicated. b-d Histograms showing expression of lilli (b), Su(Tpl) (c), and Cdk9 (d) transcripts based on normalized read counts (linear scale from DESeq2 [62]) across the gene models after zero (0d), one (1d), or two (2d) days of induction. Biological replicate numbers (#) are indicated. Values for females (left) and males (right) are shown in each panel. Knockdown expression levels following induction of lilli RNAi, Su(Tpl) RNAi, and Cdk9 DN (black), and shams (yellow or gray) as well as one standard deviation (bars) are shown. Significant differential expression (p adj < 0.05 from DESeq2) relative to sex and timepoint matched shams are shown (asterisk)

We were concerned that interfering with these important transcriptional regulators would result in cell lethality and an uninformative signature of dying cells even if we bypassed developmental defects. To assess the effect of these genes on adult viability, we shifted 3 to 4 day old adult flies from the permissive temperature of 20 °C to the restrictive temperature of 29 °C to induce transgene expression. We observed no overt lethality after up to a week. The lack of adult lethality allowed us to measure the effect of Cdk9, lilli, and Su(Tpl) on the adult steady-state transcriptome in the absence of developmental defects or reduced viability. We crossed Gal4 and Gal80 ts to the experimental lines and to two control lines, which acted as shams to monitor any differential expression due to Gal4 and Gal80 ts alone (Fig. 1a). We used these sham genotypes without UAS transgenes as controls in addition to the temperature shifts. We collected sexed samples zero, one, and two days post temperature shift to 29 °C. Flies were the same age post-eclosion (7 days) when harvested. Because germline gene expression is not controlled in the expression system we used [61], we removed the gonads from the flies before preparing polyA+ RNA for stranded RNA-seq expression profiling. All experiments were biologically triplicated to quadruplicated using three flies per sample. We used a total of 99 samples, after confirming that mismatched replicate numbers did not impact conclusions (by bootstrapping, see Methods). We obtained above background RNA-seq reads for more than 8000 genes in each sample. We used the DESeq2 [62] Normalized Read Count statistical model to measure differential expression (see Methods). For each experimental sample, we made ratiometric measurements comparing replicate experimental samples to replicate sham samples as a reference. See Methods and GEO [63] (GSE77492) for detailed protocols, data, and replication statistics. A summary of all gene expression data can be found in the Additional file 1.

We first assayed the effectiveness of the adult induction experiments (Fig. 1b to d). We detected expression of Gal4 and Gal80 ts in all samples. Prior to the temperature shift, we observed no significant differential expression in lilli relative to shams (Fig. 1b), but we did observe modest but significant (Wald test, p adj < 0.05 is used throughout) reductions in target gene expression in the Su(Tpl) RNAi flies (Fig. 1c). We also observed modest but significant overexpression of Cdk9 DN (Fig. 1d), and we confirmed that the increased expression was from the transgenic Cdk9 DN allele by measuring the fraction of the reads bearing the codon substitution (~84% at 20 °C). These data indicate that the Su(Tpl) RNAi and Cdk9 DN transgenes were not fully “off” at 20 °C. Following the temperature shift to 29 °C, we observed dramatic reduction of Su(Tpl) (Fig. 1c) and lilli (Fig. 1b) steady-state expression in the appropriate RNAi bearing flies and dramatic overexpression of Cdk9 DN (~96% from the DN allele; Fig. 1d) relative to shams. These data indicate that we altered the expression levels (and the encoded activity in the case of Cdk9) of the three genes under study.

Effects on gene expression

To explore the transcriptional responses to transgene induction, we compared replicated results from experimental samples relative to each of the two types of sham samples and asked for significantly differentially expressed genes in any of these time- and temperature-matched comparisons. We observed extensive and significant differences in the gene expression profiles relative to sham controls in these 36 pairwise comparisons, especially for reduced expression in the experimental samples (e.g., Fig. 2a, b). Overall we observed that 4792 genes showed significant differential expression in at least one pairwise comparison. This constituted 27% of all genes in the genome and 58% of the genes that we scored as expressed in all samples. To identify patterns that we could use to classify the responses of this large set of differentially expressed genes in a way that would allow us to infer which elongation complexes might be affected, we used k-means to cluster the expression ratios of experimental relative to sham samples. We removed clusters driven by background genotypes (see Methods). We observed that 579 differentially expressed genes were clearly attributed to lilli RNAi, Su(Tpl) RNAi, or Cdk9 DN (Fig. 2c) treatments. There were four clusters of genes that were dominated by reduced gene expression on treatment: cluster 1 was down-regulated by lilli RNAi, Su(Tpl) RNAi and Cdk9 DN, clusters 2 and 3 were down-regulated following lilli RNAi or Su(Tpl) RNAi (cluster 3 genes were often up-regulated in Cdk9 DN flies), and cluster 4 was down-regulated only following Cdk9 DN expression. There was strong concordance between the lilli RNAi and Su(Tpl) RNAi profiles in all clusters. That there were no clear clusters of genes differentially expressed due to only one of these RNAi treatments, suggests that wild-type Lilli and Su(Tpl) positively regulate (directly or indirectly) many of the same genes. This is consistent with the hypothesis that these proteins are often obligatory components of the same functional complex. Cluster 1 showed reduced expression following Cdk9 DN induction, suggesting that wild-type lilli, Su(Tpl), and Cdk9 positively regulate these genes (directly or indirectly), consistent with the participation of all three activities at a subset of genes. This is the correlation expected for disrupted SEC or EAP. Most gene expression altered by lilli and Su(Tpl) (cluster 2 and 3) were not regulated by Cdk9, which is consistent with the composition of MLL. The Cdk9 DN expression profile was distinct (cluster 4). Therefore, there is also Cdk9-specific differential expression profile, consistent with a role for P-TEFb alone.

Fig. 2
figure 2

Differential expression and clustering. a-b Vocano-plots showing examples of differential expression for lilli RNAi (a) and Su(Tpl) RNAi (b) one day after induction in female samples relative to Sham2 versus significance (truncated at > 75). Genes with significant differential expression (DESeq2 p adj < 0.05, red) and no significant differential expression (DESeq2 p adj ≥ 0.05, black) are shown. Gene expression are shown as log2ratios of medians (X tilde) of experimental (exp) replicates divided by sham replicates. c Differential expression heatmap of experimental samples relative to shams. Values indicated in the key (bottom) were truncated at +/- 2. Reduced (blue) and increased (red) expression in experimental samples relative to sham are shown. 579 genes (rows) with coherent differential expression relative to shams are shown. Sample comparisons (columns) relative to Sham1 (yellow fill) or Sham2 (gray fill) are indicated (bottom), as are female (f) and male (m) sex, timepoint in day(s) (d), and tested transgenes. The k-means cluster ID used in the text is shown (right) and highlighted within the heatmap (open black boxes)

Expression of genes encoding enzymes

To help determine if positive regulators of transcriptional elongation affect specific tissues or organs, we used FlyAtlas data from adults [64] to determine if differentially expressed genes had enriched expression in particular tissues. We found that the overall expression profile was not strongly enriched for genes expressed in any adult tissues (leftmost column of Fig. 3a), consistent with our use of the entire fly other than the gonads. Strikingly, the genes showing differential expression following lilli RNAi, Su(Tpl) RNAi, or Cdk9 DN induction showed coherence in implied tissue-specificity (Fig. 3a). We found that all clusters with differentially expressed genes had enriched expression in the midgut. This is intriguing, as the midgut environment is subject to rapid changes as food passes through the digestive tract.

Fig. 3
figure 3

Characteristics of differentially expressed genes. a Heatmap showing enrichment for genes expressed in tissues (rows) of wild-type flies from FlyAtlas among genes in the Fig. 2 clusters or among all genes expressed in our study (columns). The proportion of genes in a column enriched in each tissue (see key) is shown in hierarchical order. Tissues are written out except: ta ganglion = thoracicoabdominal ganglion; CNS = central nervous system; acc gland = male accessory gland; V = virgin, and M = mated. The carcass is the thorax and abdomen without gut and reproductive tract. b Heatmap showing GO biological process terms (rows) enriched in any cluster (see key). GO terms are also presented in hierarchical clustering order. c-d Browser views showing expression of two sets of related genes that are genomic neighbors and are positively regulated by lilli and Su(Tpl) (female, one day after induction). c The lysosomal α-mannosidase (LMan) genes. d The Jonah (Jon) chymotrypsin genes. Chromosome arm and genome coordinates are shown (top). The longest annotated transcript of each gene is shown. In the gene models, untranslated (gray boxes) and coding (yellow boxes) regions, and introns (gray lines) are shown. Transcription direction is shown (bent arrows). Base-level read density per million reads (RPM) (see Methods; arbitrary scale) is shown for RNAi (light green) and sham (dark green) as are test genes or sham abbreviations (right). Instances of significant induction-dependent differential expression are indicated (asterisks). e Boxplot of primary transcript length (in base pairs) by cluster (blue) or among all expressed genes (green). Clusters with significantly shorter (Wilcoxon rank test, p < 0.05) primary transcript than all expressed genes are indicated (asterisks)

To determine if there was a functional category of genes differentially expressed due to the transgenes, we performed Gene Ontology (GO) term analysis (Fig. 3b) using expressed genes as the reference, and selected all terms that were enriched in one of the clusters. The enriched terms were associated with proteolysis, various aspects of amino acid or carbohydrate metabolism, transport, and pheromones. The genes regulated by all three of the transgenes (clusters 1) were enriched for genes encoding typsins with serine-type endopeptidase function (e.g., Serine protease 6). The genes down-regulated following lilli RNAi or Su(Tpl) RNAi but not Cdk9 DN induction (clusters 2 and 3) were also enriched in genes encoding proteolysis functions in addition to metabolic, sensory, and transport functions. We show two examples of gene groups encoding important digestive enzymes that were down-regulated by transgene induction: the Lysosomal alpha-mannosidase (LMan) genes (Fig. 3c) and chymotrypsin encoding Jonah genes (Fig. 3d). We found that these genes required lilli and Su(Tpl) (but not Cdk9) for high level expression (clusters 2 and 3). They also both showed strongly midgut-biased expression in wild-type flies [64], and their wild-type functions involve carbohydrate hydrolysis in mannose production [65] and proteolysis (serine-type endopeptidase activity) [66] respectively.

We noticed that many of these enzyme-encoding genes with differential expression had short primary transcripts. Therefore, we determined if there was an overall tendency for preferential regulation of genes with short transcripts, by plotting the distributions of annotated primary transcript length of genes in each expression ratio cluster (Fig. 3e). Interestingly, the genes regulated by all three tested genes (cluster 1, see Fig. 2c) have significantly shorter primary transcripts than all expressed genes (1517 versus 2217 bp; Wilcoxon rank test, p = 4 × 10−6). Moreover, the genes regulated by lilli and Su(Tpl) (cluster 2 and 3, see Fig. 2c) also have significantly shorter primary transcripts than all expressed genes (1679 and 1027 versus 2217 bp; Wilcoxon rank test, p ≤ 8 × 10−6). In contrast, the genes regulated by Cdk9 had insignificantly shorter primary transcript length compared to all expressed genes (2099 versus 2217 bp; Wilcoxon rank test, p = 0.6). It is worth noting that the two clusters with the shortest primary transcripts (cluster 1 and 3) also had the strongest down regulation of expression after induction (Fig. 2c), as well as concordant enrichment of genes with proteolysis function (Fig. 3b). Given that control by elongation and short transcript length both contribute to rapid production of mature transcripts, these data suggest that many genes involved in metabolism have been optimized for rapid elongation.

Sex-biased regulation

We were intrigued by the role for positive elongation factors in the expression of a large number of metabolic enzyme encoding genes expressed in the midgut, as the Drosophila midgut shows sex-biased physiology and morphology, due to factors such as the immense metabolic requirements to support egg development and the requirements for specific saturation states in lipids used for pheromone production [67,68,69]. Additionally, lilli and Su(Tpl) are required for external sexual morphology [51]. This raised the possibility that there could be different roles for positive elongation factors in the sexes. To explore this hypothesis, we measured sex-biased responses to gene expression treatments relative to shams for all 4792 differentially expressed genes. We then followed a k-means clustering procedure to identify genes with sex-biased patterns (Fig. 2c). We found three clusters (514 genes in total) showing a coherent sex difference in expression (Fig. 4a). Genes in cluster I showed higher expression in males relative to females following Cdk9 DN treatment, suggesting a role for P-TEFb. Genes in cluster II showed reduced expression in males relative to females, while those in cluster III showed higher expression in males relative to females following lilli RNAi and Su(Tpl) RNAi treatments, but not Cdk9 DN. These results suggest a role for MLL in regulating sex-biased expression.

Fig. 4
figure 4

Sex-biased regulation after knockdown of elongation. a Heatmap of induction-dependent sex-biased effects. Clusters are labeled (Roman numerals). A cluster with eight genes is included in cluster II due to its small size and similar pattern as cluster II. See Fig. 2 for labeling. b Heatmap of enrichment for tissue specific expression in the clusters. See Fig. 3 for labeling, except m = male and f = female in the key. c Heatmap of GO biological process terms enriched in any of the clusters. See Fig. 3 for labeling. d-e Browser views of two examples of sex-biased gene regulation (one day after induction) are shown - CG31091 (a possible lipase encoding gene) and CG13091 (a possible fatty acyl-CoA reductase encoding gene). Note that these two CG names differ by placement of a single digit. See Fig. 3 for labeling, except that we distinguish female (red) and male (blue) expression

To determine where these genes with sex-biased responses might be expressed, we again compared to the FlyAtlas dataset (Fig. 4b). In addition to the digestive system, genes expressed in carcass were enriched in all three clusters. The FlyAtlas definition of carcass is the thorax and abdomen after removal of the other tissues and organs, so this category includes epidermis, musculature, and oenocytes, which perform liver-like functions in Drosophila [70]. We also found modest enrichment of genes expressed in a variety of non-gonadal tissues in the other clusters. To help determine the function of the genes showing sex-biased differential expression, we performed GO term enrichment analysis and observed that the most significant enrichment was trans-membrane transport and fatty acid elongation in genes regulated by Cdk9 (cluster I, Fig. 4c). Genes showing sex-biased expression due to lilli RNAi and Su(Tpl) RNAi treatment were enriched in secondary metabolic processes (cluster III, Fig. 4c). Despite the lack of significant enrichment for GO terms in cluster II, we did observe sex-biased differential expression for at least a few genes encoding metabolic functions. For example, CG31091 encodes a protein with similarity to lipases and requires lilli and Su(Tpl) activity for its male-biased expression (cluster II, Fig. 4d). This possible lipase shows highly midgut-biased expression in wild-type flies [64]. As another example, CG13091 encodes a protein with similarity to fatty acyl-CoA reductases and low level of expression in females requires lilli and Su(Tpl) (cluster II, Fig. 4e). The putative reductase is highly expressed in wild-type fat body, heart, and carcass but not in other tissues [64]. Thus, like the overall pattern of differential expression, the sex-biased response to elongation factors was enriched in genes encoding metabolic functions.

Discussion

Complex components

Various positive elongation factor complexes have been described. Many of those complexes have overlapping components and complex composition is variable in the literature (Table 1) [25, 27, 31]. Therefore, genetic data can be useful for helping to determine the constituent components in biologically active complexes, or in epistatic relationships. For example, SEC contains both Lilli and Su(Tpl), while LEC and AEP contain either Su(Tpl) or Lilli (Table 1). In our expression profiles, lilli and Su(Tpl) positively regulate a nearly identical set of genes, which is consistent with them being obligatory co-factors in MLL, SEC, and EAP complexes. Our data does not support a role for LEC or AEP, although we cannot rule out the existence of these complexes. We also found that about 20% of the genes regulated by lilli and Su(Tpl) were also regulated by Cdk9. This is consistent with the presence of lilli, Su(Tpl), and Cdk9 in SEC and EAP [71]. However, we also found hundreds of genes positively regulated by Cdk9 only. The genes specifically regulated by Cdk9 might be dependent on P-TEFb independently of the larger order complexes.

Metabolic responses

Promoter-proximal pausing of RNAPII is an interesting regulatory mechanism because it allows for rapid responses. RNAPII binding to the TSS, unwinding the DNA duplex and initiating transcription requires much more time in vivo (10 min [72, 73]) than elongation (about 1.5 k bases (kb) per minute [74, 75]). Such rapid responses are especially important for heat shock, pathogen defense, and proper expression of some developmentally regulated genes [36, 39, 44, 45]. We found that positive elongation factors regulate many genes encoding metabolic enzymes, especially those expressed in the midgut. Release from pausing is an attractive mechanism for rapidly producing enzymes in response to feeding. Indeed, the Jonah genes encode chymotrypsins in the midgut and are expressed in feeding, but not in non-feeding stages of development, such as the first 12 h of adult life when flies are maturing and using residual fat body stores [76]. In mosquitos, the orthologous genes are rapidly induced in the gut by a blood meal [77, 78]. We suggest that transcriptional elongation is an important regulator of rapid metabolic gene expression responses in adult Drosophila.

Rapid transcription also depends on gene length. Interestingly, genes positively regulated by the lilli and Su(Tpl) genes in our studies tend to have short transcripts, especially the ones with the highest level of differential expression after perturbation. For instance, most of the transcripts of chemotrypsin genes, such as Jonah genes and lambdaTry, are shorter than 1 kb, which enables transcription in less than 40 s. In general, the genes positively regulated by the lilli and Su(Tpl) genes produce even shorter transcripts than the heat shock genes (1751 versus 2231 bp). Although it may not be a prerequisite, short transcript length could be a feature evolutionarily favored by genes with rapid responses. We did not observe significantly shorter transcripts among genes regulated by Cdk9, suggesting that regulation of short transcripts is MLL-dependent.

Sex-biased expression

The lilli and Su(Tpl) loci contain many conserved and highly occupied Dsx binding sites, and driving lilli RNAi or Su(Tpl) RNAi in dsx expressing cells results in specific switches in sexual dimorphism in the male first leg and the female genitalia, suggesting that these genes are required in both females and males, but in different locations [51]. A sex-biased role for Cdk9 has not been described, but the locus does have a conserved consensus Dsx binding site [51]. However, we do not know if this sex- and tissue-specific role is confined to morphology, or is more general. We found that sex-biased expression changed for about 200 genes due to lilli RNAi or Su(Tpl) RNAi, and for about 300 genes due to Cdk9 DN . As was the case for morphology, the lilli and Su(Tpl) genes are required for both genes with female- and male-biased expression. This is consistent with the idea that sex-biased expression is highly context dependent and argues against strictly female- and male-biased batteries of genes that shows the same bias in all tissues. The Cdk9 regulated genes showed a more directional effect of reduced expression in females relative to males.

Many of the genes regulated by elongation factors encode metabolic functions. There is a great deal of precedent for sex-biased metabolism. For example, egg production is energy intensive and tightly linked to nutrient availability via insulin signaling [79]. At the morphological level, the Drosophila midgut is nearly twice as long in females as males and is regulated by the sexual identity pathway [67]. Finally, lipids and other metabolites also show dramatic sexual dimorphism [80]. Our work suggests that some of the sex-biased gene expression in metabolic pathways is due to indirect activity of Dsx target genes such as lilli and Su(Tpl).

Conclusions

Turning down positive elongation factors has a global transcriptional effect. Our data suggest that Lilli and Su(Tpl) are likely to be obligatory cofactors for the expression of many genes in soma, and that Cdk9 has both shared and Lilli/Su(Tpl)-independent functions. Our data also suggest that transcriptional elongation regulation by these factors is essential for rapid enzymatic and metabolic activities during digestion, a novel category of function regulated by rapid transcriptional elongation. In addition, the differential expression in females versus males suggests that positive elongation factors play roles in sex-biased gene regulation.

Methods

Summarized data is found in Additional file 1.  A table of key resources is provided in Additional file 2, which describes the reagent or resources used; including chemical and biological reagents, critical commercial kits, genotypes of the flies, deposited data, and software. We also provided the sources and identifiers for these resources.

Molecular genetics

Cdk9 DN transgenic lines were generated by PCR-amplifying the Cdk9 open reading frame from w 1118 genomic DNA and cloned into pBluescript KS. Mutant Cdk9 DN, a substitution of amino acid 199 of the active site (aspartic acid to asparagine) was obtained by the megaprimer method [81] using a mutagenic oligonucleotide. The mutant DNA was cloned in the UASp vector [82] and injected in w 1118 embryos. A transgenic line with an unmapped insertion on the 3rd chromosome was used in this study, but other lines show the same effect on development in the presence of various Gal4 drivers.

Flies

All flies were reared on the following media (per liter: 6.2 g agar, 59 g cornmeal, 59 g yeast, 5 ml propionic acid and 26 ml 99% methyl 4-hydroxybenzoate). Briefly, homozygous w*; +; P{tubP-GAL4} LL7, P{tubP-GAL80 ts}7 virgin females were crossed to homozygous males with a UAS responsive transgene or homozygous males of control lines for shams. The paternal lines were lilli RNAi, Su(Tpl) RNAi, Cdk9 DN, w 1118, and lacZ. There are additional experiments in the GEO [63] series accession GSE77492 not presented here. First, we performed profiling following gpp RNAi but did not shown them in the results due to poor knockdown. Second, we used flies with an additional copy of Cdk9 DN, but observed a non-additive effect of two copies of Cdk9 DN. Third, we ectopically expressed stand still, but these samples were for other purposes. All the parental lines were reared and crossed at 20 °C, which also applied to their progeny. We collected adult progeny at 0 to 4 h post-eclosion, aged them for 5 to 7 days in fresh vials, and shifted to the restrictive temperature (29 °C) for 0, 1 or 2 day(s). All the flies were 7 days old at the time of dissection.

RNA-seq

For each sample, three gonadectomized adults were dissected and crushed in 100 ul RNAlater solution (Life Technologies, Carlsbad CA) before sealing in deep 96-well plates at room temperature. Samples were homogenized with a mini-beadbeater (Biospec Products, Bartlesville OK) three times for 60 s each in the presence of 20 to 30 1.0 mm glass beads per sample (BioSpec Products, Bartlesville OK). We extracted RNA using RNeasy according to the manufacturer (Qiagen, Valencia CA), except that we added 600 ul RLT buffer and 700 ul 70% ethanol in each well. We used 400 ng total RNA in 50 ul nuclease-free water (Quality Biological, Gaithersburg MD) for RNA-seq library preparation. We strictly followed a previously reported polyA+ strand-specific RNA-seq library preparation protocol [83]. We conducted single-end 50 bp RNA-seq on the HiSeq2000 Sequencing System (Illumina, San Diego CA). Reads were mapped to FlyBase [84] release 6.04 using STAR (v2.4.2a) with default parameters [85]. The overall agreement between biological replicates was ≥ 0.88 (Pearson’s r). We also mapped the reads of all samples to the intergenic regions, which we used as a measure of background expression noise for calculating the number of expressed genes. Genes expressed at < 95% percentile of intergenic expression were counted as unexpressed [86]. All samples with 29 °C treatments were triplicated, and all samples without 29 °C treatments were quadruplicated except for female Sham2 samples (triplicated). To confirm that our results were robust to mismatched sample sizes, we used bootstrapping (100x) to randomly select three replicates among each sample set. The median number of re-identified differentially expressed genes was 96%, suggesting that the variable samples sizes have little impact on our results. Therefore, we used all replicates in this work.

Expression analysis

For each sample, uniquely mapped read counts from HTSeq (v0.6.1p1) (--stranded = reverse) [87] were used as inputs in DESeq2 (v1.10.1). We used Normalized Read Counts as the unit of expression except for genome browser coverage plots, where we used base-level coverage of reads (generated by bedtools v2.25.0 [88]) normalized by RNA-seq library size in millions (RPM). For Normalized Read Counts, DESeq2 calculates the geometric mean (GM) of each sample, and divides the raw read counts by the GM to obtain ratios of relative expression. For each sample, we used the median ratio as a sample-specific size factor (~1 in our experiments), and normalized read counts were calculated as raw read counts divided by the size factor. Previous studies based 726 Drosophila individuals [86] demonstrated that the DESeq2 method was a high performing normalization approach relative to total counts, upper quartile, median, trimmed mean of M-values, quantile, reads per kilobase per million mapped reads, and un-normalized count data. We also used DESeq2 to calculate differential expression. For each experimental sample within the same sex and treatment, we compared experimental relative to each of the two sham genotypes. We retained genes with significantly differential expression (Wald test p adj < 0.05) in any of the comparisons. We further categorized these genes by their induction-dependent differential expression (log2 ratio of the experiment expression relative to sham) across samples. We converted “N/A” values (divide by 0) to “0” (indicating no bias) for k-means clustering. We used the parameter k = 18 (Fig. 2c) or 20 (Fig. 4a) based on evaluation of within-cluster sum of squares [89], and also confirmed that the major clusters were robust from k = 15 to 25. We used visual examination to remove clusters driven by differences between the two sham genotypes and clusters that showed weak differential expression, or responded even without induction.

We used FlyAtlas [64] microarray expression data to infer where differentially expressed genes were expressed in our experiments. We converted microarray Oligo IDs to FlyBase [84] gene IDs using DAVID (v6.7) [90]. Some genes have multiple microarray probes, and we calculated the mean signal using 1 = “enriched”, 0 = “not different”, and -1 = “depleted” as compared to the signal in the whole body. We defined enriched expression as mean > 0. Gene Ontology (GO) [91] tests were conducted in FlyMine [92]. We used a Holm-Bonferroni cutoff of p adj < 0.05 for GO analysis and all tests were based on the background of expressed genes in any of our samples rather than all genes. We used the pheatmap package (v1.0.8) in R (v3.2.2) to generate all heat maps of tissue specificity and GO results.

Abbreviations

AEP:

AF4 family/ENL family/P-TEFb complex

AF4:

ALL1-Fused Gene From Chromosome 4

AFF4:

AF4/FMR2 family member 4

Cdk9 :

Cyclin-dependent kinase 9

CTD:

Carboxy-terminal domain of RNAPII

DN:

Dominant negative

DotCom:

Dot1 complex

DRB:

5,6-dichloro-1-β-D-ribofuranosylbenzimidazole

DSIF:

DRB sensitivity inducing factor

EAP:

Elongation assisting proteins

FMR2:

Fragile mental retardation 2

GM:

Geometric mean

GO:

Gene ontology

Gpp :

grappa

H3K79:

Histone H3 Lys 79

LEC:

Little Elongation Complex

Lilli :

lilliputian

LMan :

Lysosomal alpha-mannosidase

MLL:

Mixed-lineage leukemia 1 proteins

NELF:

Negative elongation factor

P-TEFb:

Positive transcription elongation factor b complex

RNAPII:

RNA polymerase II

SEC:

Super Elongation Complex

Su(Tpl) :

Suppressor of Triplolethal

TSS:

Transcription start site

References

  1. Myer VE, Young RA. RNA polymerase II holoenzymes and subcomplexes. J Biol Chem. 1998;273:27757–60.

    Article  CAS  PubMed  Google Scholar 

  2. Young RA. RNA polymerase II. Annu Rev Biochem. 1991;60:689–715.

    Article  CAS  PubMed  Google Scholar 

  3. Vannini A, Cramer P. Conservation between the RNA polymerase I, II, and III transcription initiation machineries. Mol Cell. 2012;45:439–46.

    Article  CAS  PubMed  Google Scholar 

  4. Revyakin A, Liu C, Ebright RH, Strick TR. Abortive initiation and productive initiation by RNA polymerase involve DNA scrunching. Science. 2006;314:1139–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jonkers I, Lis JT. Getting up to speed with transcription elongation by RNA polymerase II. Nat Rev Mol Cell Biol. 2015;16:167–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. C Quaresma AJ, Bugai A, Barboric M. Cracking the control of RNA polymerase II elongation by 7SK snRNP and P-TEFb. Nucleic Acids Res. 2016;44:7527–39.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Min IM, Waterfall JJ, Core LJ, Munroe RJ, Schimenti J, Lis JT. Regulating RNA polymerase pausing and transcription elongation in embryonic stem cells. Genes Dev. 2011;25:742–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yamaguchi Y, Takagi T, Wada T, Yano K, Furuya A, Sugimoto S, Hasegawa J, Handa H. NELF, a multisubunit complex containing RD, cooperates with DSIF to repress RNA polymerase II elongation. Cell. 1999;97:41–51.

    Article  CAS  PubMed  Google Scholar 

  9. Missra A, Gilmour DS. Interactions between DSIF (DRB sensitivity inducing factor), NELF (negative elongation factor), and the Drosophila RNA polymerase II transcription elongation complex. Proc Natl Acad Sci U S A. 2010;107:11301–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Adelman K, Lis JT. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nat Rev Genet. 2012;13:720–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wada T, Takagi T, Yamaguchi Y, Ferdous A, Imai T, Hirose S, Sugimoto S, Yano K, Hartzog GA, Winston F, et al. DSIF, a novel transcription elongation factor that regulates RNA polymerase II processivity, is composed of human Spt4 and Spt5 homologs. Genes Dev. 1998;12:343–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wenzel S, Schweimer K, Rosch P, Wohrl BM. The small hSpt4 subunit of the human transcription elongation factor DSIF is a Zn-finger protein with alpha/beta type topology. Biochem Biophys Res Commun. 2008;370:414–8.

    Article  CAS  PubMed  Google Scholar 

  13. Nonet M, Sweetser D, Young RA. Functional redundancy and structural polymorphism in the large subunit of RNA polymerase II. Cell. 1987;50:909–15.

    Article  CAS  PubMed  Google Scholar 

  14. Zehring WA, Lee JM, Weeks JR, Jokerst RS, Greenleaf AL. The C-terminal repeat domain of RNA polymerase II largest subunit is essential in vivo but is not required for accurate transcription initiation in vitro. Proc Natl Acad Sci U S A. 1988;85:3698–702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Phatnani HP, Greenleaf AL. Phosphorylation and functions of the RNA polymerase II CTD. Genes Dev. 2006;20:2922–36.

    Article  CAS  PubMed  Google Scholar 

  16. Ping YH, Rana TM. DSIF and NELF interact with RNA polymerase II elongation complex and HIV-1 Tat stimulates P-TEFb-mediated phosphorylation of RNA polymerase II and DSIF during transcription elongation. J Biol Chem. 2001;276:12951–8.

    Article  CAS  PubMed  Google Scholar 

  17. Peterlin BM, Price DH. Controlling the elongation phase of transcription with P-TEFb. Mol Cell. 2006;23:297–305.

    Article  CAS  PubMed  Google Scholar 

  18. Mancebo HS, Lee G, Flygare J, Tomassini J, Luu P, Zhu Y, Peng J, Blau C, Hazuda D, Price D, Flores O. P-TEFb kinase is required for HIV Tat transcriptional activation in vivo and in vitro. Genes Dev. 1997;11:2633–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yang X, Gold MO, Tang DN, Lewis DE, Aguilar-Cordova E, Rice AP, Herrmann CH. TAK, an HIV Tat-associated kinase, is a member of the cyclin-dependent family of protein kinases and is induced by activation of peripheral blood lymphocytes and differentiation of promonocytic cell lines. Proc Natl Acad Sci U S A. 1997;94:12331–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhu Y, Pe’ery T, Peng J, Ramanathan Y, Marshall N, Marshall T, Amendt B, Mathews MB, Price DH. Transcription elongation factor P-TEFb is required for HIV-1 tat transactivation in vitro. Genes Dev. 1997;11:2622–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yu DS, Zhao R, Hsu EL, Cayer J, Ye F, Guo Y, Shyr Y, Cortez D. Cyclin-dependent kinase 9-cyclin K functions in the replication stress response. EMBO Rep. 2010;11:876–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Smith E, Lin C, Shilatifard A. The super elongation complex (SEC) and MLL in development and disease. Genes Dev. 2011;25:661–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lin C, Smith ER, Takahashi H, Lai KC, Martin-Brown S, Florens L, Washburn MP, Conaway JW, Conaway RC, Shilatifard A. AFF4, a component of the ELL/P-TEFb elongation complex and a shared subunit of MLL chimeras, can link transcription elongation to leukemia. Mol Cell. 2010;37:429–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Luo Z, Lin C, Shilatifard A. The super elongation complex (SEC) family in transcriptional control. Nat Rev Mol Cell Biol. 2012;13:543–7.

    Article  CAS  PubMed  Google Scholar 

  25. Chou S, Upton H, Bao K, Schulze-Gahmen U, Samelson AJ, He N, Nowak A, Lu H, Krogan NJ, Zhou Q, Alber T. HIV-1 Tat recruits transcription elongation factors dispersed along a flexible AFF4 scaffold. Proc Natl Acad Sci U S A. 2013;110:E123–31.

    Article  CAS  PubMed  Google Scholar 

  26. He N, Liu M, Hsu J, Xue Y, Chou S, Burlingame A, Krogan NJ, Alber T, Zhou Q. HIV-1 Tat and host AFF4 recruit two transcription elongation factors into a bifunctional complex for coordinated activation of HIV-1 transcription. Mol Cell. 2010;38:428–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nguyen AT, Zhang Y. The diverse functions of Dot1 and H3K79 methylation. Genes Dev. 2011;25:1345–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shanower GA, Muller M, Blanton JL, Honti V, Gyurkovics H, Schedl P. Characterization of the grappa gene, the Drosophila histone H3 lysine 79 methyltransferase. Genetics. 2005;169:173–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Farooq Z, Banday S, Pandita TK, Altaf M. The many faces of histone H3K79 methylation. Mutat Res Rev Mutat Res. 2016;768:46–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kim W, Choi M, Kim JE. The histone methyltransferase Dot1/DOT1L as a critical regulator of the cell cycle. Cell Cycle. 2014;13:726–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Biswas D, Milne TA, Basrur V, Kim J, Elenitoba-Johnson KS, Allis CD, Roeder RG. Function of leukemogenic mixed lineage leukemia 1 (MLL) fusion proteins through distinct partner protein complexes. Proc Natl Acad Sci U S A. 2011;108:15751–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bonifer C, Cockerill PN. Transcriptional and Epigenetic Mechanisms Regulating Normal and Aberrant Blood Cell Development Preface. Transcriptional and Epigenetic Mechanisms Regulating Normal and Aberrant Blood Cell Development 2014;V-Vii.

  33. Smith ER, Lin C, Garrett AS, Thornton J, Mohaghegh N, Hu D, Jackson J, Saraf A, Swanson SK, Seidel C, et al. The little elongation complex regulates small nuclear RNA transcription. Mol Cell. 2011;44:954–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yokoyama A, Lin M, Naresh A, Kitabayashi I, Cleary ML. A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription. Cancer Cell. 2010;17:198–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Core LJ, Lis JT. Transcription regulation through promoter-proximal pausing of RNA polymerase II. Science. 2008;319:1791–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Muse GW, Gilchrist DA, Nechaev S, Shah R, Parker JS, Grissom SF, Zeitlinger J, Adelman K. RNA polymerase is poised for activation across the genome. Nat Genet. 2007;39:1507–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Smith E, Shilatifard A. Transcriptional elongation checkpoint control in development and disease. Genes Dev. 2013;27:1079–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zeitlinger J, Stark A, Kellis M, Hong JW, Nechaev S, Adelman K, Levine M, Young RA. RNA polymerase stalling at developmental control genes in the Drosophila melanogaster embryo. Nat Genet. 2007;39:1512–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Saunders A, Core LJ, Lis JT. Breaking barriers to transcription elongation. Nat Rev Mol Cell Biol. 2006;7:557–67.

    Article  CAS  PubMed  Google Scholar 

  40. Conaway JW, Shilatifard A, Dvir A, Conaway RC. Control of elongation by RNA polymerase II. Trends Biochem Sci. 2000;25:375–80.

    Article  CAS  PubMed  Google Scholar 

  41. Stetina T, Kostal V, Korbelova J. The Role of Inducible Hsp70, and Other Heat Shock Proteins, in Adaptive Complex of Cold Tolerance of the Fruit Fly (Drosophila melanogaster). PLoS One. 2015;10, e0128976.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lis JT, Mason P, Peng J, Price DH, Werner J. P-TEFb kinase recruitment and function at heat shock loci. Genes Dev. 2000;14:792–803.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Shim EY, Walker AK, Shi Y, Blackwell TK. CDK-9/cyclin T (P-TEFb) is required in two postinitiation pathways for transcription in the C. elegans embryo. Genes Dev. 2002;16:2135–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xu J, Grant G, Sabin LR, Gordesky-Gold B, Yasunaga A, Tudor M, Cherry S. Transcriptional pausing controls a rapid antiviral innate immune response in Drosophila. Cell Host Microbe. 2012;12:531–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang X, Lee C, Gilmour DS, Gergen JP. Transcription elongation controls cell fate specification in the Drosophila embryo. Genes Dev. 2007;21:1031–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Erickson JW, Quintero JJ. Indirect effects of ploidy suggest X chromosome dose, not the X:A ratio, signals sex in Drosophila. PLoS Biol. 2007;5, e332.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Lucchesi JC, Kuroda MI. Dosage compensation in Drosophila. Cold Spring Harb Perspect Biol. 2015;7.

  48. Hempel LU, Oliver B. Sex-specific DoublesexM expression in subsets of Drosophila somatic gonad cells. BMC Dev Biol. 2007;7:113.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Manoli DS, Foss M, Villella A, Taylor BJ, Hall JC, Baker BS. Male-specific fruitless specifies the neural substrates of Drosophila courtship behaviour. Nature. 2005;436:395–400.

    CAS  PubMed  Google Scholar 

  50. Siwicki KK, Kravitz EA. Fruitless, doublesex and the genetics of social behavior in Drosophila melanogaster. Curr Opin Neurobiol. 2009;19:200–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Clough E, Jimenez E, Kim YA, Whitworth C, Neville MC, Hempel LU, Pavlou HJ, Chen ZX, Sturgill D, Dale RK, et al. Sex- and tissue-specific functions of Drosophila doublesex transcription factor target genes. Dev Cell. 2014;31:761–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Picard MA, Cosseau C, Mouahid G, Duval D, Grunau C, Toulza E, Allienne JF, Boissier J. The roles of Dmrt (Double sex/Male-abnormal-3 Related Transcription factor) genes in sex determination and differentiation mechanisms: Ubiquity and diversity across the animal kingdom. C R Biol. 2015;338:451–62.

    Article  PubMed  Google Scholar 

  53. Luo SD, Shi GW, Baker BS. Direct targets of the D. melanogaster DSXF protein and the evolution of sexual development. Development. 2011;138:2761–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mummery-Widmer JL, Yamazaki M, Stoeger T, Novatchkova M, Bhalerao S, Chen D, Dietzl G, Dickson BJ, Knoblich JA. Genome-wide analysis of Notch signalling in Drosophila by transgenic RNAi. Nature. 2009;458:987–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Luschnig S, Moussian B, Krauss J, Desjeux I, Perkovic J, Nusslein-Volhard C. An F1 genetic screen for maternal-effect mutations affecting embryonic pattern formation in Drosophila melanogaster. Genetics. 2004;167:325–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Brand AH, Perrimon N. Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development. 1993;118:401–15.

    CAS  PubMed  Google Scholar 

  57. Ferris J, Ge H, Liu L, Roman G. G(o) signaling is required for Drosophila associative learning. Nat Neurosci. 2006;9:1036–40.

    Article  CAS  PubMed  Google Scholar 

  58. Perkins LA, Holderbaum L, Tao R, Hu Y, Sopko R, McCall K, Yang-Zhou D, Flockhart I, Binari R, Shim HS, et al. The Transgenic RNAi Project at Harvard Medical School: Resources and Validation. Genetics. 2015;201:843–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Garriga J, Segura E, Mayol X, Grubmeyer C, Grana X. Phosphorylation site specificity of the CDC2-related kinase PITALRE. Biochem J. 1996;320(Pt 3):983–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Peng J, Marshall NF, Price DH. Identification of a cyclin subunit required for the function of Drosophila P-TEFb. J Biol Chem. 1998;273:13855–60.

    Article  CAS  PubMed  Google Scholar 

  61. Rorth P. Gal4 in the Drosophila female germline. Mech Dev. 1998;78:113–8.

    Article  CAS  PubMed  Google Scholar 

  62. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chintapalli VR, Wang J, Dow JA. Using FlyAtlas to identify better Drosophila melanogaster models of human disease. Nat Genet. 2007;39:715–20.

    Article  CAS  PubMed  Google Scholar 

  65. Chng WB, Bou Sleiman MS, Schupfer F, Lemaitre B. Transforming growth factor beta/activin signaling functions as a sugar-sensing feedback loop to regulate digestive enzyme expression. Cell Rep. 2014;9:336–48.

    Article  CAS  PubMed  Google Scholar 

  66. Ross J, Jiang H, Kanost MR, Wang Y. Serine proteases and their homologs in the Drosophila melanogaster genome: an initial analysis of sequence conservation and phylogenetic relationships. Gene. 2003;304:117–31.

    Article  CAS  PubMed  Google Scholar 

  67. Hudry B, Khadayate S, Miguel-Aliaga I. The sexual identity of adult intestinal stem cells controls organ size and plasticity. Nature. 2016;530:344–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Reiff T, Jacobson J, Cognigni P, Antonello Z, Ballesta E, Tan KJ, Yew JY, Dominguez M, Miguel-Aliaga I. Endocrine remodelling of the adult intestine sustains reproduction in Drosophila. Elife. 2015;4, e06930.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Wicker-Thomas C, Henriet C, Dallerac R. Partial characterization of a fatty acid desaturase gene in Drosophila melanogaster. Insect Biochem Mol Biol. 1997;27:963–72.

    Article  CAS  PubMed  Google Scholar 

  70. Gutierrez E, Wiggins D, Fielding B, Gould AP. Specialized hepatocyte-like cells regulate Drosophila lipid metabolism. Nature. 2007;445:275–80.

    Article  CAS  PubMed  Google Scholar 

  71. Murali T, Pacifico S, Yu J, Guest S, Roberts 3rd GG, Finley Jr RL. DroID 2011: a comprehensive, integrated resource for protein, transcription factor, RNA and gene interactions for Drosophila. Nucleic Acids Res. 2011;39:D736–43.

    Article  CAS  PubMed  Google Scholar 

  72. Lis J. Promoter-associated pausing in promoter architecture and postinitiation transcriptional regulation. Cold Spring Harb Symp Quant Biol. 1998;63:347–56.

    Article  CAS  PubMed  Google Scholar 

  73. Adelman K, Marr MT, Werner J, Saunders A, Ni Z, Andrulis ED, Lis JT. Efficient release from promoter-proximal stall sites requires transcript cleavage factor TFIIS. Mol Cell. 2005;17:103–12.

    Article  CAS  PubMed  Google Scholar 

  74. Yao J, Ardehali MB, Fecko CJ, Webb WW, Lis JT. Intranuclear distribution and local dynamics of RNA polymerase II during transcription activation. Mol Cell. 2007;28:978–90.

    Article  CAS  PubMed  Google Scholar 

  75. Ardehali MB, Lis JT. Tracking rates of transcription and splicing in vivo. Nat Struct Mol Biol. 2009;16:1123–4.

    Article  CAS  PubMed  Google Scholar 

  76. Carlson JR, Hogness DS. Developmental and functional analysis of Jonah gene expression. Dev Biol. 1985;108:355–68.

    Article  CAS  PubMed  Google Scholar 

  77. Edwards MJ, Lemos FJ, Donnelly-Doman M, Jacobs-Lorena M. Rapid induction by a blood meal of a carboxypeptidase gene in the gut of the mosquito Anopheles gambiae. Insect Biochem Mol Biol. 1997;27:1063–72.

    Article  CAS  PubMed  Google Scholar 

  78. Muller HM, Crampton JM, della Torre A, Sinden R, Crisanti A. Members of a trypsin gene family in Anopheles gambiae are induced in the gut by blood meal. EMBO J. 1993;12:2891–900.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Ikeya T, Galic M, Belawat P, Nairz K, Hafen E. Nutrient-dependent expression of insulin-like peptides from neuroendocrine cells in the CNS contributes to growth regulation in Drosophila. Curr Biol. 2002;12:1293–300.

    Article  CAS  PubMed  Google Scholar 

  80. Parisi M, Li R, Oliver B. Lipid profiles of female and male Drosophila. BMC Res Notes. 2011;4:198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Kammann M, Laufs J, Schell J, Gronenborn B. Rapid insertional mutagenesis of DNA by polymerase chain reaction (PCR). Nucleic Acids Res. 1989;17:5404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Thummel CS, Boulet AM, Lipshitz HD. Vectors for Drosophila P-element-mediated transformation and tissue culture transfection. Gene. 1988;74:445–56.

    Article  CAS  PubMed  Google Scholar 

  83. Lee H, Cho DY, Whitworth C, Eisman R, Phelps M, Roote J, Kaufman T, Cook K, Russell S, Przytycka T, Oliver B. Effects of Gene Dose, Chromatin, and Network Topology on Expression in Drosophila melanogaster. PLoS Genet. 2016;12, e1006295.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Gramates LS, Marygold SJ, Santos GD, Urbano JM, Antonazzo G, Matthews BB, Rey AJ, Tabone CJ, Crosby MA, Emmert DB, et al. FlyBase at 25: looking to the future. Nucleic Acids Res. 2017;45:D663–71.

    Article  PubMed  Google Scholar 

  85. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.

    Article  CAS  PubMed  Google Scholar 

  86. Lin Y, Golovnina K, Chen ZX, Lee HN, Negron YL, Sultana H, Oliver B, Harbison ST. Comparison of normalization and differential expression analyses using RNA-Seq data from 726 individual Drosophila melanogaster. BMC Genomics. 2016;17:28.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Anders S, Pyl PT, Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–9.

    Article  CAS  PubMed  Google Scholar 

  88. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Witten DM, Tibshirani R. A framework for feature selection in clustering. J Am Stat Assoc. 2010;105:713–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.

    Article  CAS  Google Scholar 

  91. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lyne R, Smith R, Rutherford K, Wakeling M, Varley A, Guillier F, Janssens H, Ji W, McLaren P, North P, et al. FlyMine: an integrated database for Drosophila and Anopheles genomics. Genome Biol. 2007;8:R129.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Kohoutek J. P-TEFb- the final frontier. Cell Div. 2009;4:19.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Lin X, Taube R, Fujinaga K, Peterlin BM. P-TEFb containing cyclin K and Cdk9 can activate transcription via RNA. J Biol Chem. 2002;277:16873–8.

    Article  CAS  PubMed  Google Scholar 

  95. Mohan M, Herz HM, Takahashi YH, Lin C, Lai KC, Zhang Y, Washburn MP, Florens L, Shilatifard A. Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom). Genes Dev. 2010;24:574–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mueller D, Bach C, Zeisig D, Garcia-Cuellar MP, Monroe S, Sreekumar A, Zhou R, Nesvizhskii A, Chinnaiyan A, Hess JL, Slany RK. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood. 2007;110:4445–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Rowley JD. The critical role of chromosome translocations in human leukemias. Annu Rev Genet. 1998;32:495–519.

    Article  CAS  PubMed  Google Scholar 

  98. Tenney K, Shilatifard A. A COMPASS in the voyage of defining the role of trithorax/MLL-containing complexes: linking leukemogensis to covalent modifications of chromatin. J Cell Biochem. 2005;95:429–36.

    Article  CAS  PubMed  Google Scholar 

  99. Huret JL, Dessen P, Bernheim A. An atlas of chromosomes in hematological malignancies. Example: 11q23 and MLL partners. Leukemia. 2001;15:987–9.

    Article  CAS  PubMed  Google Scholar 

  100. Dahlberg O, Shilkova O, Tang M, Holmqvist PH, Mannervik M. P-TEFb, the super elongation complex and mediator regulate a subset of non-paused genes during early Drosophila embryo development. PLoS Genet. 2015;11, e1004971.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to Cale Whitworth and Hangnoh Lee for help with RNA-seq library preparation, and Harold Smith and the NIDDK Genomics core for sequencing. This work utilized the computational resources of the NIH High-Performance Computing (HPC) Biowulf cluster (http://hpc.nih.gov). We would also like to thank Emily Clough for discussion and suggestions. This research was supported in part by the Intramural Research Program of the NIH, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK).

Funding

This research was supported in part by the Intramural Research Program of the NIH, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK).

Availability of data and materials

GEO series accession (GSE77492).

Authors’ contributions

DP and BO designed the experiment. DB, DP, and BO performed fly work and HY performed the genomics. HY and BO analyzed data and wrote the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Haiwang Yang.

Additional files

Additional file 1:

Summary of gene expression and k-means clustering. (XLS 10240 kb)

Additional file 2:

Key resources table. (XLS 55 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, H., Basquin, D., Pauli, D. et al. Drosophila melanogaster positive transcriptional elongation factors regulate metabolic and sex-biased expression in adults. BMC Genomics 18, 384 (2017). https://doi.org/10.1186/s12864-017-3755-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-017-3755-x

Keywords