Skip to main content
Advertisement
  • Loading metrics

Trans-regulatory changes underpin the evolution of the Drosophila immune response

Abstract

When an animal is infected, the expression of a large suite of genes is changed, resulting in an immune response that can defend the host. Despite much evidence that the sequence of proteins in the immune system can evolve rapidly, the evolution of gene expression is comparatively poorly understood. We therefore investigated the transcriptional response to parasitoid wasp infection in Drosophila simulans and D. sechellia. Although these species are closely related, there has been a large scale divergence in the expression of immune-responsive genes in their two main immune tissues, the fat body and hemocytes. Many genes, including those encoding molecules that directly kill pathogens, have cis regulatory changes, frequently resulting in large differences in their expression in the two species. However, these changes in cis regulation overwhelmingly affected gene expression in immune-challenged and uninfected animals alike. Divergence in the response to infection was controlled in trans. We argue that altering trans-regulatory factors, such as signalling pathways or immune modulators, may allow natural selection to alter the expression of large numbers of immune-responsive genes in a coordinated fashion.

Author summary

A fundamental question in biology is the nature of the genetic changes underlying evolutionary change, and immune systems provide an ideal system to examine this as they tend to evolve fast as animals adapt to an ever-changing array of parasites and pathogens. Comparing two species of the fruit fly Drosophila, we found that the transcriptional response to infection evolves extremely fast. However, changes in cis (where the genetic change is on the same DNA molecule as the gene in question) and trans (where the genetic change can be elsewhere in the genome) are playing different roles. Changes in cis frequently caused large differences in immune gene expression between species, but these differences were seen regardless of whether the animal was infected. In contrast, changes in trans were responsible for altering how gene expression changes in response to infection. Immune responses are complex and multifaceted, requiring the expression of many genes to be altered in a tightly regulated manner when the animal is infected. Natural selection acting on trans regulatory factors may allow the expression of many downstream genes to be altered in a coordinated fashion.

Introduction

Pathogens and parasites drive rapid evolutionary change in their hosts, and it is common to find closely related species which differ greatly in their susceptibility to infection [1]. Drosophila, like most animals, relies on an innate immune system to defend itself against infection, where immune receptors and effectors are encoded in the germline. Infection is detected when receptors bind to non-self, such as peptidoglycan from bacterial cells or molecules derived from parasitic wasps [2]. Alongside this, the immune system can sense damage to host tissues, such as when a parasitic wasp ovipositor pierces the cuticle [3]. These processes activate signalling pathways, including the Toll and Imd pathways which culminate in NF-kappaB transcription factors translocating into the nucleus [4]. This results in a rapid transcriptional response [5]. Humoral immunity centres on the fat body, which secretes molecules such as antimicrobial peptides into circulation [4]. Meanwhile, immune cells called hemocytes (blood cells) proliferate, move into circulation, and differentiate into specialised forms [6]. These cells can phagocytize microbes, and produce the enzymes required for the synthesis of melanin, which can encapsulate pathogens and parasites. Together, these defences provide a powerful protection against infection.

Innate immune systems evolve rapidly. For example, in Drosophila, natural selection has driven the rapid divergence of the peptide sequence of immune-related genes [79]. However, despite changes in gene expression underpinning the immune response, it remains poorly understood how patterns of immune gene expression change during evolution. The expression of a specific immune effector such as an antimicrobial peptide could be changed in two ways: by altering cis-regulatory elements (CRE), which are DNA sequences that regulate the expression of nearby genes (e.g., promoters, enhancers), or upstream factors that control the expression of the gene in trans, such as pathogen recognition molecules, signalling pathways, transcription factors or immune modulators. A fundamental question is whether evolutionary change tends to occur when gene expression is altered in cis or trans. Work on a variety of species has found that cis-regulatory divergence between species is extremely common, affecting a large proportion of genes across genomes [1014]. However, these cis-regulatory changes tend to be constant across environments, and differences in how gene expression responds to the environment are controlled in trans [13, 15, 16]. In this study we have investigated whether these principles also govern the evolution of gene regulation during the immune response.

In Drosophila, there is evidence that variation in both cis- and trans-acting factors controlling the expression of immune effectors may affect susceptibility to infection. Genetic polymorphisms in regulatory proteins, such as signalling pathway components, affect susceptibility [17], suggesting these may affect downstream effectors in trans. Furthermore, these interact with non-coding variants in antimicrobial peptide genes [17], suggesting a role for cis regulatory variation in effector genes. The role of cis- and trans-regulatory polymorphisms in controlling the expression of immunity genes in the fat body has been investigated in two lines of D. melanogaster that were infected with bacteria [18]. In uninfected flies, cis-regulatory polymorphisms were the dominant cause of differences in the expression of immune-responsive genes. This was also the case when flies were infected with Gram-positive Enterococcus faecalis, but when they were infected with Gram-negative Serratia marcescens differences in expression were mainly controlled in trans [18]. Together with its well-understood immune system, these observations mean Drosophila provides the opportunity to understand how regulation of genes expression changes during the evolution of immune systems.

To understand the changes to gene regulation that underlie the evolution of immune responses, we have compared two closely related species where an ecological shift is thought to have caused recent and rapid divergence in their immune defences. Parasitoid wasps are common parasites of Drosophila, laying their eggs in fly larvae and eventually killing their hosts. When a Drosophila melanogaster larva is parasitised, the fat body secretes humoral immune factors into circulation, while specialised hemocytes called lamellocytes differentiate [19]. If the immune response is successful, these cells encapsulate the parasitoid egg and undergo melanisation, killing the parasitoid [20]. The Drosophila simulans immune response against parasitoids closely resembles that seen in D. melanogaster. However, Drosophila sechellia is thought to have escaped from parasitoid attack by feeding on a fruit that is toxic to the wasps [21]. Following this ecological shift, it has evolved a greatly altered transcriptional response to parasitoid attack [22], produces few lamellocytes and is unable to encapsulate parasitoids [23, 24]. As these changes have occurred within the last few hundred thousand years [25], we have used these species as a model to understand the evolution of the transcriptional response to infection.

We used RNA sequencing to distinguish the cis and trans regulatory changes underlying the differing immune responses of these species. While differences in gene expression between the two species can be caused by both cis and trans divergence, in F1 hybrids alleles inherited from both parental species are within the same cellular environment and therefore under identical trans control. Therefore, the relative abundance of transcripts from the two alleles of a gene in F1 hybrids captures the cis regulatory differences between the two species [26, 27]. This means that by comparing the expression levels of the two alleles in hybrids (cis) to the total regulatory difference between parental species (cis+trans), we can estimate the extent of divergence in trans regulation [10, 14]. This approach has been applied to several closely related pairs of species, frequently with the result indicating that differences in gene expression are predominantly the result of cis differences [1014]. Here, we have extended this approach to understand how cis and trans regulatory changes have led to the divergence of the immune responses of D. simulans and D. sechellia.

Results

D. sechellia has lost the ability to encapsulate

The Drosophila immune system kills parasitoid wasp eggs by encapsulating them in a layer of immune cells called hemocytes and then melanising this cellular capsule. To characterise how the susceptibility of D. simulans and D. sechellia to parasitoid wasp infection differed, we allowed L. boulardi to oviposit in the fly larvae and dissected them after 96 hours. In line with previous reports [23, 24], we found that D. sechellia never encapsulated the wasp eggs but D. simulans can mount an encapsulation response (Fig 1A).

thumbnail
Fig 1. The immune response of two species of Drosophila and their hybrids.

(A) The proportion of larvae that had encapsulated eggs of the parasitoid wasp L. boulardi 96 hours post infection (96hpi). (B and C). The proportion of encapsulated droplets of mineral oil containing wasp homogenate in (B) larvae 24 hours post injection or (C) in adult flies that developed from injected larva. Error bars are 95% binomial confidence intervals.

https://doi.org/10.1371/journal.pgen.1010453.g001

Parasitoid wasps can suppress and evade the immune response. To investigate the immune response without any countermeasures from the parasitoid, we injected the larvae with a droplet of mineral oil that contains homogenised parasitoid tissue [2] and examined the rate the oil droplet was encapsulated (Fig 1). D. simulans had encapsulated 6.6% of oil droplets 24 hours post injection (hpi), which is a time-point when Drosophila larvae are encapsulating wasp eggs [28]. By the time adults developed from injected larvae, 67% of oil droplets were encapsulated. In contrast, D. sechellia is unable to mount an encapsulation response, as even when flies had developed into adults none of the oil droplets had been encapsulated. Unexpectedly, F1 hybrids encapsulated at higher rates than either parent, largely due to them mounting a faster response than D. simulans—90.2% of oil droplets were encapsulated in larvae and 98.8% in emerged flies (Fig 1).

The cellular and humoral immune response of D. simulans and D. sechellia has extensively diverged

To understand the immunological reasons why the two species differ in susceptibility to infection, we examined the transcriptional response to immune challenge in the two main immune tissues of Drosophila—hemocytes and the fat body. We injected larvae with droplets of mineral oil containing wasp homogenate and measured gene expression by RNAseq 24 hours later. This time-point was selected as it is when parasitoids are being encapsulated [28], and there is a large transcriptional response to infection [29]. After filtering out low expression genes, there were 4969 and 5275 genes remaining in fat body and hemocyte data-sets respectively. Of these, 531 genes in fat body and 1971 genes in hemocytes exhibited significant (FDR < 0.05) expression changes after immune challenge in at least one species or F1 hybrids.

In hemocytes, many more genes were differentially expressed in D. simulans than D. sechellia (FDR<0.05; coloured points in Fig 2A). Furthermore, different genes are changing their expression following infection in the two species, as there is only a weak correlation between their transcriptional response (Fig 2B; Pearson’s correlation of data in this figure: ρ = 0.06, P<0.007, 95% CI [0.02, 0.10]; S4 Dataset).

thumbnail
Fig 2. Divergence of the transcriptional response to immune challenge in fat body and hemocytes.

Larvae were immune challenged by injecting them with mineral oil containing wasp homogenate while control larvae were not injected. The transcriptional response was then measured by RNA sequencing 24 hours post injection. (A) Volcano plots comparing the magnitude and statistical significance of changes in gene expression after immune challenge. Each point is a gene, and they are coloured above an FDR threshold of 5%. (B) The magnitude of the transcriptional response to immune challenge in D. simulans compared to D. sechellia. Each point is a gene.

https://doi.org/10.1371/journal.pgen.1010453.g002

The cellular immune response to parasitoid wasps involves the differentiation of specialised immune cells called lamellocytes [30]. Using single-cell RNA sequencing, we have described how cells differentiate through immature forms (LAM1 and LAM2) to mature lamellocytes (LAM3) [31]. Using the data from that study, we used digital cytometry [32] to deconvolute our bulk RNA-seq data and infer the proportion of hemocytes in these different transcriptional states (Fig 3A). Despite D. sechellia producing few lamellocytes after infection, we unexpectedly inferred that mature and immature lamellocytes were present in uninfected D. sechellia larvae. The abundance of these cells only slightly increased after immune challenge. The presence of cells whose transcriptome resembles lamellocytes in D. sechellia is surprising as we know from studies of cell morphology that these cells are not produced in this species [23]. However, our previous work in D. melanogaster has shown that some genotypes of flies produce cells which transcriptionally resemble lamellocytes but only adopt the classical lamellocyte morphology after infection [31]. This suggests that the changes to the hemocyte population that have occurred in D. sechellia are more complex than simply losing the ability to differentiate lamellocytes.

thumbnail
Fig 3. Changes in gene expression after immune challenge.

(A) Proportion of lamellocytes in different transcriptional states in D. simulans and D. sechellia larvae under control and immune-challenged conditions, estimated from bulk RNA-seq data with CIBERSORTx. Error bars are 95% confidence intervals. (B) Changes in the expression of immunity genes in the D. simulans and D. sechellia fat body after immune challenge. Bars represent the scaled mean count per million of gene expression in control larvae and 24 hours after injection with mineral oil containing wasp homogenate. Only genes with a greater than two-fold change in expression in at least one species are shown. Significance of the change in expression after infection was defined by false discovery rate (FDR): 0.05*, 0.01**, 0.001***.

https://doi.org/10.1371/journal.pgen.1010453.g003

In D. simulans there were few mature or immature lamellocytes before infection, but these cell states became abundant after immune challenge (Fig 3A). This contrasts with only small shifts in the cell population in D.sechellia Therefore, the larger transcriptional response of D. simulans hemocytes to immune challenge is likely caused by the differentiation of lamellocytes and their precursors.

In the fat body, there is weak correlation in the transcriptional response of the two species to immune challenge (Fig 2B; Pearson’s correlation in data in plot: ρ = 0.09, P<0.05, 95% CI [0.01, 0.17]; S4 Dataset), suggesting that the humoral immune response of the two species largely involves different genes. When looking at genes with large increases in expression after immune challenge, the difference between the two species is less stark than was the case for hemocytes, although the largest fold changes in expression were seen in D. simulans (Fig 2A).

The fat body plays a key role in the humoral immune response by secreting a diverse range of immune-related molecules into the hemolymph. We identified 84 differentially expressed genes with a log2 fold change greater than two in at least one species (FDR<0.05). As expected, a gene ontology term analysis found that these were enriched in genes involved in the defence response (P<0.05, Holm-Bonferroni corrected). A key role of the fat body is to secrete antimicrobial peptides (AMPs), and several of these were up-regulated in both species (Fig 3B). Bomanins are essential in killing some bacterial pathogens [33, 34], and they were significantly up-regulated upon immune challenge in D. sechellia but not D. simulans (Fig 3B). The opposite is the case for three genes encoding Turandots, which are stress response genes that are commonly induced after infection [35]. The expression of these genes increased approximately 1000-fold in D. simulans, but they were not significantly up-regulated in D. sechellia (Fig 3B). In most cases the differences in fold-change-after-infection between the species was not significant, but in the final section of the results we measure the expression of a selection of effector genes by quantitative PCR in whole larvae, and this confirmed that AMPs and Bomanins are more highly expressed in D. sechellia than D. simulans. Together, our results demonstrate that the humoral immune response of these closely related species to parasitoids has diverged considerably.

Trans-regulatory factors underlie divergence in the cellular immune response

Our data on allele-specific expression in hybrids allowed us to examine the relative importance of cis and trans regulatory changes in causing evolutionary divergence in the cellular immune response. Here, we restricted our analysis to the 1971 genes that were differentially regulated in hemocytes following immune challenge (Fig 4).

thumbnail
Fig 4. Expression divergence of immune responsive genes between D. simulans and D. sechellia in hemocytes.

Plots only show genes that are significantly differentially expressed in response to infection in at least one species. (A) Expression differences between parental species (total regulatory divergence) plotted against allelic expression differences within F1 hybrids (cis-regulatory divergence). Gene expression was measured under unchallenged control conditions (left) and immune-challenged conditions (centre), and this was used to estimate the expression change in response to immune challenge (right). Each point is a gene, the expression difference of each gene is represented as the relative log2 fold change. (A and B) The genes are colour coded according to whether gene expression divergence is controlled in cis or in trans, with panel (B) depicting the number of genes in each category. The ambiguous category refers to genes with significant divergence in expression between the parental species but no significant divergence in either cis or trans. (C) Histograms depicting the frequencies of cis- and trans-diverged genes across the magnitudes of total expression divergence between D. simulans and D. sechellia. Fly larvae were immune-challenged by injecting them with oil droplets containing wasp homogenate.

https://doi.org/10.1371/journal.pgen.1010453.g004

First, we examined the immune response—the magnitude by which expression changed following an immune challenge (Fig 4, right column). We found no evidence of any genes where the response to infection had diverged due to changes in cis (Fig 4A and 4B, right column; no genes where cis effect with FDR<0.05). In contrast, changes in trans altered the response to infection in 659 genes (Fig 4A and 4B, right column; FDR<0.05). Furthermore, the magnitude of these effects was frequently large, with changes in trans causing the response of some genes to differ by 1000-fold or more between D. simulans and D. sechellia (Fig 4C, right column). Therefore, differences in the cellular immune response of the two species are caused by trans regulatory divergence, likely controlling the differentiation of immune cells after infection (Fig 3A).

Next, we examined the expression of these immune-responsive genes under control conditions, where the immune response has not been activated. Here the pattern was dramatically different. As well as many immune-responsive genes with trans-regulatory divergence (732 of 1971 genes, 37.1%; FDR<0.05), there was also extensive cis-regulatory divergence (663 of 1971, 33.6%; FDR<0.05) (Fig 4A and 4B, left column). The divergence of both cis regulatory elements and trans regulatory factors was frequently associated with very large changes in gene expression, sometimes exceeding a 100-fold change in mRNA levels. However, there was a tendency for trans effects to cause larger changes in gene expression than cis effects (Fig 4C). To assess the overall importance of cis and trans effects, we examined the correlation between the total divergence in gene expression between the parental species and divergence in cis or trans. This revealed that total changes in gene expression were more strongly correlated with the trans-regulatory divergence (ρ = 0.73, 95% CI [0.71, 0.75], Pearson’s correlation of log2 fold change of gene expression across 1971 genes, S6 Dataset) than cis-regulatory divergence (ρ = 0.54, 95% CI [0.51, 0.57], S6 Dataset). A similar pattern emerges when looking at the expression of genes in the hemocytes of immune-challenged larvae. Here, the number of immune responsive genes that diverged under trans-control decreased (Fig 4B). However, the total gene expression divergence between D. simulans and D. sechellia remained strongly correlated with both trans- (ρ = 0.70, 95% CI [0.68, 0.73], S6 Dataset) and cis-divergence (ρ = 0.66, 95% CI [0.63, 0.68], S6 Dataset), due to the trans-regulatory changes leading to larger expression differences (Fig 4C). Overall, this data demonstrates that in both infected and uninfected larvae, divergence in the expression of immune responsive genes is caused by changes in both cis and trans. However, while changes in cis affect expression in both infected and uninfected larvae alike, changes in trans frequently alter how expression responds to infection.

Dysregulation of cellular immunity in hybrids

To understand how regulatory factors of the two species interact, we examined the transcriptional response to immune challenge in hybrids. As the response in hemocytes is driven by the differentiation of lamellocytes, we initially examined the expression of atilla, which is specifically expressed in mature lamellocytes and commonly used to identify this cell type [36]. In uninfected F1 hybrids, the expression of atilla [36] was overdominant, exceeding the expression in D. simulans by 3.6 times and D. sechellia by 4.3 times. This is consistent with the immune response in hybrids becoming dysregulated, such that this cell type or its precursors are present even in uninfected individuals. After immune-challenge, the expression of atilla increased in D. simulans but not D. sechellia. This resulted in atilla expression in D. simulans and hybrids being respectively 32 and 16 times that in D. sechellia (p < 10−6, quasi-likelihood F-test). This is consistent with the observation that few lamellocytes differentiate in D. sechellia.

These observations indicate that hybrids already possess activated hemocytes pre-infection, and the immune challenge in D. simulans causes it to ‘catch up’ with hybrids. To explore this further, we used digital cytometry (see above) to estimate the proportion of hemocytes in different transcriptional states in hybrids. As was the case in D. sechellia, lamellocytes or their precursors were present before immune challenge. However, after immune challenge the number of these cells increase (S5 Fig). Therefore the hybrids combine features of both parents—activation of these cells before infection (D. sechellia) and induction after infection (D. simulans). As a consequence of these cell states being present before immune challenge, the magnitude of the transcriptional response to infection in hybrid larvae was far less than in D. simulans (Fig 2A).

There was also a substantial number of other genes that were misexpressed in the hemocytes of hybrids, with expression levels sometimes deviating strongly from levels in both the parental species (S4 Fig). In unchallenged larvae, this was apparent in a group of genes that had lower expression in the hybrids than either parent, a pattern known as underdominance (S4(A) Fig, left column). After immune challenge, the expression of many of these genes was restored to normal levels (S4(A) Fig, centre column). This increase in gene expression after infection means that their response to immune challenge was overdominant (S4(B) Fig, right column). Other genes showed dominance of the D. sechellia allele resulting from the expression of lamellocyte markers described above (S4(B) Fig).

Trans-regulatory changes drive divergence in the humoral immune response

Unlike hemocytes, the humoral immune response in the fat body is not known to involve the differentiation of new cell types, but instead leads to cells secreting molecules into the haemolymph. As was the case for hemocytes, we focused our analysis on genes that were differentially expressed in at least one of the species after immune challenge. In our control flies where the immune response has not been activated, the expression of 46% of these genes had diverged in cis (244 of 531 immune responsive genes; Fig 5A and 5B). In contrast, only 15.6% of the genes had significant divergence in trans (83 genes; Fig 5A and 5B). As a consequence, the total divergence of gene expression between the two species was more strongly correlated with cis-regulatory divergence than trans-regulatory divergence (cis: Pearson’s ρ = 0.73, 95% CI [0.69, 0.77]; trans: ρ = 0.55, 95% CI [0.49, 0.61]). Therefore, in uninfected larvae, differences in the expression of immune-responsive genes are predominantly caused by changes to cis regulatory elements.

thumbnail
Fig 5. Expression divergence of immune responsive genes between D. simulans and D. sechellia in the fat body.

Plots only show genes that are significantly differentially expressed in response to infection in at least one species. (A) Expression differences between parental species plotted against allelic expression differences within F1 hybrids (cis-regulatory divergence). Gene expression was measured under unchallenged control conditions (left) and immune-challenged conditions (centre), and this was used to estimate the expression change in response to immune challenge (right). (A and B) The genes are colour coded according to whether gene expression divergence is controlled in cis or in trans, with panel (B) depicting the number of genes in each category. (C) Histograms depicting the frequencies of cis- and trans-diverged genes with differing expression divergence between D. simulans and D. sechellia. (D) Changes in the expression of immunity genes encoding components of major humoral immune response signalling pathways in the fat body after immune challenge. Bars represent the change in gene expression 24 hours after injection with mineral oil containing wasp homogenate. Significantly differentially expressed genes within each species were indicated by * on top of columns. Divergent gene expression change responding to immune challenge between D. simulans and D. sechellia was indicated by * on top of bars between columns. Significance level was defined by false discovery rate (FDR): 0.05*, 0.01**, 0.001***.

https://doi.org/10.1371/journal.pgen.1010453.g005

In contrast to the situation in uninfected larvae, divergence in the response to immune-challenge between the two species is driven by changes to trans regulatory factors. Examining the fold-change in gene expression after immune activation, we did not find any genes where there were significant cis regulatory differences, but 31 genes had significant divergence in trans (Fig 5A and 5B, right-hand column). The importance of trans effects is reflected in a strong correlation between the divergence in the response of these genes to immune-challenge and the trans-effect (Fig 5) (ρ = 0.93, 95% CI [0.91, 0.94], Pearson’s correlation).

These analyses demonstrate that while cis regulatory divergence is the dominant factor causing differences in the expression of humoral immune genes in uninfected flies, changes in the response to infection is controlled in trans. The combination of these two effects is apparent when comparing levels of gene expression in immune-challenged larva of the two species. Here, the number of immune-responsive genes with trans-regulatory divergence has increased compared to unchallenged control conditions (Fig 5B, centre plot), as has the correlation between the divergence in gene expression and trans-regulatory divergence (ρ = 0.65, 95% CI [0.60, 0.70], Pearson’s correlation).

The humoral immune response in the fat body is regulated in a large part by the immune deficiency (Imd), Toll and JAK-STAT pathways, often acting in combination. For example, Baramicin A and Bomanin genes are regulated by the Toll pathway [3739], while Cecropin A, Attacin A, Defensin and Metchnikowin are regulated to varying degrees by both Toll and Imd. Therefore, differences in the expression of Bomanin and AMP genes between D. sechellia and D. simulans suggest the Toll pathway and potentially other immune signalling pathways may be causing differences in expression in trans (Fig 3B, see below for verification by quantitative PCR). We therefore investigated whether genes encoding components of immune signalling pathways differed in their expression between the species. We found that six genes in the Toll and Imd pathways, including those encoding the NF-κB transcription factors Dif and Relish, were significantly up-regulated upon immune challenge in D. sechellia but not D. simulans (Fig 5D). In four of these cases the difference in immune induction was significant between the species (Fig 5D). The up-regulation of these genes in D. sechellia therefore provides a potential explanation of why Bomanins and AMPs are more highly expressed in this species.

To examine whether these pathways might underlie differences in gene expression between the species that are regulated in trans more broadly, we searched for binding site motifs of six immune related transcription factors (Relish, Dif, Dorsal, STAT, Serpent and CrebA) in the region 1kB upstream of the 531 immune responsive genes. However, we failed to find any enrichment of these motifs upstream of genes showing trans regulatory divergence (S2A Table). Similarly, there was no association between genes where only one species possessed the motif and divergence in cis (S2B Table).

To further understand how trans-regulatory factors had diverged between these species, we examined the immune response in F1 hybrid larvae. In contrast to hemocytes, the immune response of the hybrid fat body most closely resembled D. simulans. In unchallenged control fat bodies, there was no clear tendency for gene expression in hybrids to resemble either parent (S4 Fig). However, after infection the level of expression in D. simulans tended to be dominant, with changes in expression level in the hybrids being similar to D. simulans (S4(B) Fig). This is consistent with a dominant trans-acting factor inherited from D. simulans controlling the humoral immune response in hybrids.

Cis-regulatory divergence causes large changes in the expression of immune effectors

To provide increased statistical power to examine how changes to cis regulatory elements affect the transcriptional response to infection, we investigated divergence in the expression of a small number of humoral immune-response genes with greater replication. As reported previously, parasitoid exposure up-regulates antimicrobial peptides and stress response genes [29]. As many of these genes had considerable cis differences between species, we selected 13 genes that were mostly known to play a role in these responses. To allow us to increase our sample size, we used whole larvae rather than dissecting single tissues, so the results are not directly comparable to the RNAseq experiment. We designed primers to amplify targets within exons of these genes from cDNA or genomic DNA templates and sequenced the products using Illumina sequencing (S1 Table). We used SNPs within the reads to determine whether they were from the D. simulans or D. sechellia allele. When we amplified genomic DNA from F1 hybrids, approximately half the reads were from the D. simulans allele and half from the D. sechellia allele, confirming there is no PCR bias causing one of the alleles to be preferentially amplified (Fig 6A).

thumbnail
Fig 6. Cis regulatory divergence in humoral immunity genes.

Transcripts were amplified by PCR from F1 hybrids between D. simulans and D. sechellia, and the PCR products Illumina sequenced. RNA was extracted from whole larvae that were uninfected (control) or immune challenged (wasp). Genomic DNA (gDNA) was included to check there were no biases towards one allele. (A) The proportion of reads from the two alleles. (B) The natural log of the odds (logit) that a transcript is from the D. simulans allele. Error bars are 95% confidence intervals on model coefficients. Fly larvae were immune-challenged by injecting them with oil droplets containing wasp homogenate. The horizontal dashed line (y = 0.5) signifies that there is no cis-regulatory divergence between the two species.

https://doi.org/10.1371/journal.pgen.1010453.g006

There was allele-specific expression of all 13 genes in F1 hybrids, indicating that there is interspecific cis-regulatory divergence (Fig 6A and 6B). In some cases, the magnitude of these effects is large. For example, for Bomanin Bc2 (BomBc2) there were 18 times more D. sechellia transcripts than D. simulans transcripts in unchallenged larvae. Similarly, after immune challenge, there was more than 12 times more D. simulans TotB transcripts than D. sechellia transcripts (Fig 6B). These cis-regulatory differences affected several classes of immune effectors where the concentration of the gene product is expected to affect susceptibility to infection. For example, Bomanins are a family of secreted proteins induced after infection that play a central role in killing Gram-positive bacteria and fungi [33, 34]. We found that the D. simulans allele of three Bomanin genes had greater expression, but in the fourth gene the D. sechellia allele was more highly expressed (Fig 6A and 6B). The antimicrobial peptides Metchnikowin (Mtk) and Cecropin (Cec) kill fungi and Gram-negative bacteria respectively. Mtk, CecA1 and CecC all showed evidence of allele-specific expression, and again in some cases the D. simulans allele was more highly expressed, and in other cases the D. sechellia allele (Fig 6A and 6B). Similarly, we found that there was significant cis-regulatory divergence in the expression of the parasitoid-induced gene Edin [40] in both uninfected and immune-challenged larvae.

Cis- and trans-regulatory changes combine to switch TotX between inducible and constitutive expression

To investigate how the expression of these 13 humoral response genes differed between the parental species, we measured their expression by quantitative PCR. This revealed that the stress-response gene TotX is up-regulated ∼100X after immune challenge in D. simulans, but it is constitutively expressed at a high level in D. sechellia, irrespective of whether the immune response has been activated (Fig 7A). To understand the role of cis and trans divergence in this evolutionary shift, we also measured the expression of this gene in the F1 hybrids. By combining these measurements with the allele-specific expression data described above (Fig 6), we could estimate the expression level of each allele in hybrids (Fig 7A). This revealed that both the D. simulans and D. sechellia alleles are immune-induced in F1 hybrids, demonstrating that trans regulatory differences between the species are essential for the gene to be constitutively expressed in D. sechellia (Fig 7B; interaction of the parameters allele, Parents/Hybrids and immune challenge, i.e., trans regulatory effect on immune response: p < 10−5 after Bonferroni correction). Using this data to estimate the magnitude of trans-regulatory divergence between species, we found trans-acting factors cause large TotX expression differences in uninfected larvae of the two species (Fig 7B, grey points). In contrast, there was no significant trans-regulatory divergence in infected larvae (Fig 7B, black points).

thumbnail
Fig 7. Relative expression between D. simulans and D. sechellia alleles and Trans regulatory divergence in humoral immunity genes.

Relative gene expressions were quantified by qPCR in parental and F1 hybrid larvae. The relative allelic expression in hybrids were estimated using proportions inferred from Illumina MiSeq sequencing of hybrid transcriptions for each gene, and the relative allelic expression between parental larvae were calibrated to half accordingly. (A) Relative allelic expressions between parental larvae and F1 hybrids under unchallenged (control), and infected (immune challenge imposed by injections of wasp homogenate) conditions. (B) Trans-regulatory divergence between D. simulans and D. sechellia in unchallenged and infected larvae, measured as the log2 ratio of relative D. simulans over D. sechellia expression. Error bars are 95% confidence intervals on model coefficient.

https://doi.org/10.1371/journal.pgen.1010453.g007

While divergence in trans is required for TotX to switch between constitutive and induced expression, TotX was also the only gene where the direction of allele-specific expression in F1 hybrids was reversed when flies were immune challenged (Fig 6). The D. sechellia allele is more highly expressed in uninfected larvae, but the D. simulans allele is most highly expressed after immune challenge (Fig 6; GLM: p < 10−5 after Bonferroni correction). Together, this suggests that the evolutionary transition between this gene being immune-induced and constitutively expressed likely results from the interaction between changes in both cis and trans.

We were able to identify this cis-regulatory change in TotX that altered the immune response due to the greater statistical power of this dataset compared to the RNAseq data. We therefore repeated this analysis on the 12 other selected genes and found that there were three where the degree of allele-specific expression differed between uninfected and immune-challenged larvae (Fig 6; Mtk, BomBc3, CecC; GLM: p < 0.01 after Bonferroni correction). In the case of Metchnikowin, an antifungal peptide, the magnitude of allele-specific expression is reduced when flies are immune-challenged (Fig 6; GLM: p < 10−9 after Bonferroni correction). However, the other two genes had only subtle changes in allele-specific expression after infection. While these results demonstrate that cis-regultory changes can affect the response to infection, this sample of genes were hand-picked based on the RNAseq data, so it does not challenge the general conclusion from the unbiased RNAseq analysis that most divergence in the immune response is controlled in trans.

We also measured the expression of these genes by quantitative PCR and estimated the trans-regulatory divergence in the same way as for TotX. All but Indy showed significant trans regulatory divergence in unchallenged larvae, and this remained significant in eight of the genes post immune challenge (Fig 7B). It is striking that all these genes also had significant cis-regulatory divergence (Fig 6), supporting our RNAseq analysis where many genes were affected by both cis and trans divergence. The interplay between these changes is illustrated by the Bomanin genes, which encode peptides that kill bacteria and fungi. All four of these genes were most highly expressed in D. sechellia, and trans regulatory divergence increased the expression of all four genes in this species (Fig 7A). However, in three of the four genes, cis regulatory divergence had the opposite effect, with the D. sechellia allele being most highly expressed (Fig 7A). This effect is known as ‘compensation’, and may evolve if stabilising selection acts on CREs to restore gene expression levels to their original level after a trans-regulatory element has changed. In the fourth gene, BomBc2, the cis and trans effects reinforced each other (Fig 7A).

Discussion

Following an immune challenge, D. simulans and D. sechellia both exhibited large transcriptional responses, with hundreds of genes changing their expression level across the two major immune tissues, hemocytes and the fat body. However, despite these species being closely related, the nature of this transcriptional response has diverged considerably, resulting in there being little correlation between the responses of the two species. As the genes involved include key immune effector molecules and markers of immune cell differentiation, these transcriptional differences likely underpin the differences in susceptibility to infection seen between these two species. The protein sequence of Drosophila immunity genes frequently evolves rapidly [7], and these results demonstrate that the same is true for gene expression. Divergence in both cis- and trans-regulatory factors have caused large changes in the expression of many immune-responsive genes. However, changes in cis overwhelmingly altered constitutive levels of gene expression, affecting immune-challenged and uninfected animals alike. In contrast, changes in the response to infection were caused by trans-regulatory factors (Figs 4 and 5).

An important question is which pathways are causing divergence in trans. We found that a number of immune effector genes are more highly expressed in D. sechellia due to trans-regulatory divergence. A possible explanation is that genes encoding transcription factors and signalling pathway components have diverged in their expression between the two species, with components of both the Toll and Imd pathways being significantly up-regulated after infection in D. sechellia but not D. simulans. This result is reminiscent of a study of two lines of D. melanogaster where trans regulatory polymorphisms had affected the immune response to a Gram negative bacterium [18]. These lines differed in the expression of several regulators of the Imd pathway, which controls the response to Gram-negative bacteria [18]. However, when we predicted whether genes were regulated by the Toll or Imd pathways from the presence of transcription factor binding motifs, we were unable to link divergence in trans with the presence of specific motifs. We would caution however that we do not now how accurately this simple analysis predicted immune-responsive enhancers.

Immune responses are complex, requiring many genes to be differentially expressed in a coordinated fashion. Altering trans-regulatory factors may provide a mechanism by which natural selection can alter these complex responses in a coordinated fashion that would be difficult to achieve by altering the CREs of numerous effector genes. Our data cannot identify what these trans-acting factors are, but they may be involved in recognising infection, signalling or immune modulation. The nature of the changes to these upstream genes is also unknown—they could have changed in cis, trans, sequence, or been lost or duplicated.

The patterns we see may be an example of a more general phenomenon, with trans-regulatory changes underpinning the evolution of how gene expression responds to the environment [15]. For example, comparing yeast species across different media, cis divergence remains consistent across environments, while trans divergence differed between environments [13]. Similarly, in Caenorhabditis elegans, trans eQTLs were responsible for differing responses to heat stress while cis eQTLs were unspecific and shared across environments [16]. In the D. melanogaster fat body, polymorphisms in gene expression are mostly controlled in cis in uninfected flies, but in trans after S. marcescens infection (although not after E. faecalis infection). These studies mirror our observations, where we found the trans regulatory divergence driving the divergence of immune response, while changes in cis were constant across infected and uninfected animals. Trans-regulatory changes to gene expression may be environment-dependent because trans-regulatory pathways have evolved to control an organism’s plastic phenotypic response to the environment, or because mutations that alter the environmental sensitivity of CREs rarely arise.

The relative roles of cis and trans changes in evolution have long been debated by developmental biologists, who have argued that CREs play an important role in the evolution, in part because their modular structure allows them to alter expression of a single gene in specific cells at specific times [41]. In contrast, changes in trans may affect many downstream genes, causing harmful pleiotropic side effects [41]. These arguments apply to developmental patterning genes that regulate the expression of many other genes, while we are mainly studying terminal effector genes where expression changes are predicted to have fewer pleiotropic effects [42]. In this regard, immune systems may resemble other systems controlling organisms’ response to environmental stressors, such as heat shock, which tend to have many terminal effector genes and fewer regulatory genes [43]. These systems are thought to be more resilient to trans changes, due to less interconnection between trans-regulatory pathways [43].

While differences in the response to infection are overwhelmingly controlled in trans, we did find a few genes where changes in cis alter the response to infection. Over longer periods of evolutionary time, the accumulation of these cis changes may allow the nature of immune responses to change by altering the expression of specific genes. Our clearest example of cis changes altering the response to infection was in the stress response gene TotX, where the direction of allele-specific expression switched upon infection. However, in this gene there was also massive divergence in trans, and it is likely the interaction of these cis and trans changes that switched the gene between immune-induced and constitutive expression.

We activated the anti-parasitoid immune response, and parasitoids are known to drive especially rapid evolution of their hosts [23, 44]. The immune response of these species is thought to have rapidly diverged following an ecological shift where D. sechellia escaped from the parasitoids following a change in the fruit on which it feeds [21]. Therefore, the selection pressures driving this change likely reflect a relaxation of selective pressure, as opposed to directional selection as may commonly be the case during the coevolution of hosts and parasites. It therefore remains to be seen whether the patterns we observe can be generalised to immune system evolution more broadly. Nonetheless, such relaxation of selection may be common, as pathogens come and go during evolution. Furthermore, many of the genes that were differentially expressed are thought to be involved in killing microbes and not parasitoids. This broad immune response following parasitoid infection has been observed in other Drosophila spp., where there is strong up-regulation of antimicrobial peptides [1, 22, 29, 45]. These genes are likely up-regulated because our immune challenge (like parasitism) injures the fly larvae [3, 46], and because the same signalling molecules are used in different aspects of the immune response. Notably, it was recently found that extracellular serine proteases that regulate the Toll pathway also regulate the melanisation response [47]. Therefore, the divergence of the anti-parasitoid immune response and the Toll-regulated genes like Bomanins may be caused by the same molecular changes and selection pressures. Regardless of its causes, it gave us insights into the evolution of immunity beyond just factors involved in killing parasitoids.

There is a fundamental difference between the causes of changes in gene expression in the cellular and humoral immune responses. The cellular immune response relies on the differentiation of new cell types [4850], and many of the changes we see likely result from the differentiation of specialised immune cells called lamellocytes in D. simulans. This can explain why there is far greater differential gene expression following immune challenge in D. simulans than D. sechellia. In contrast to our results in hemocytes, the humoral response in the fat body does not involve the differentiation of new cell types, and here trans regulatory factors are more likely to be affecting classical immune signalling pathways controlling the immune response. Despite distinctive characteristics of humoral and cellular immunity, there is extensive cross-talk between them during immune activation. Hemocytes mediate signals to the fat body during bacterial infection, facilitating in the activation of immune pathways and induction of antimicrobial peptides released by the fat body [51, 52]. On the other hand, activation of the Toll pathway in the Drosophila larval fat body is sufficient to launch a cellular response parallel to that after parasitoid oviposition, albeit inconsequential in defence against the parasitoid wasp, Leptopilina boulardi [53]. These interactions are likely underlying many of the patterns we see, with changes in the hemocytes causing changes in gene expression in the fat body or vice versa.

In conclusion, we have found that cis- and trans-regulatory changes play fundamentally different roles during the evolution of Drosophila immunity. Changes to the response to infection almost exclusively result from changes to trans-regulatory factors, while cis regulatory elements alter the expression of specific genes in both infected and uninfected animals.

Materials and methods

Drosophila lines and larvae preparation

The Sz49 line from the D. simulans panel [54] was selected as one of the parental lines. They were first collected in Zuma beach, California in 2012 and went through 15 generations of full sib matings from a single-mated female. We confirmed they were capable of encapsulating Leptopilina boulardi (strain G486) eggs prior to the experiment. The D. sechellia line DenisNF100 was from an isofemale line first collected from Denis Island in the Seychelles in 2012 [55], and subsequently sequenced [56].

D. simulans (Sz49) [54] and D. sechellia (DenisNF100) [56] were reared at 25°C with a 12-hour light/12-hour dark cycle. Virgin female flies were collected within 4 hours of emergence from the pupa and kept in vials for 2 days to confirm they did not lay fertile eggs. Mesh netting was placed over 50 mm Petri dishes containing standard cornmeal Drosophila food [57] with flies to limit space and encourage mating. To further encourage cross-species mating, twenty D. simulans virgin females were crossed with forty D. sechellia males in each Petri dish to produce F1 hybrids. Twenty D. simulans of each sex were used in each Petri dish for larvae production. To account for low egg production in D. sechellia, thirty flies of either sex were used in each Petri dish for larvae production. Ten Petri dishes were set up for each species and F1 hybrid.

Second instar female larvae were collected from each species and the hybrids 48 hours after hatching and assigned either to control groups with no treatment, or to being injected with wasp homogenate to induce an immune reaction. To produce the wasp homogenate, twenty male L. boulardi were placed on ice and homogenised in 200 μL of mineral oil with plastic pestles. For immune inductions, 4.6 nL of wasp homogenate was injected using a Nanoject II micro-injector (Drummond scientific, Bromall, PA, USA) into each larva towards the posterior end.

Calculating encapsulation rate

To quantify the encapsulation rates of D. simulans and D. sechellia against wasp eggs, twenty second instar larvae were collected from each species and put in a cornmeal vial with 2 female L. boulardi for 3 hours in 18°C. After oviposition, the wasps were taken out of the vials and the infected fly larvae were left in 25°C for 96 hours before encapsulation assay. At 96 hours post infection (hpi), infected larvae were collected and washed in double-distilled water. Oviposition was confirmed by dissecting the infected larvae with tweezers under microscope to reveal either a wasp larva or a melanised egg. The encapsulation rate was calculated by dividing the total number of the fly larvae with a confirmed wasp oviposition by the number of fly larvae with an encapsulated wasp egg. For encapsulation assay in fly larvae against wasp homogenate, injected larvae were collected at 24 hpi and washed in double-distilled water before inspection for capsules by eye under microscope. For encapsulation assay in adult flies against wasp homogenate, the flies emerged from injected larvae were collected then anaesthetised by carbon dioxide and squashed between 2 microscope slides before placed under the microscope for visual inspection for capsules. Encapsulation rate was calculated as dividing the total number of larvae or fly by the number of larvae or fly that, through visual assessment, encapsulated the wasp homogenate droplet.

Library preparation for RNA sequencing

At 24 hpi, groups of 70 larvae were bled for hemocytes by tearing the cuticle from the ventral side with tweezers, and groups of 10 larvae were dissected for fat bodies for each tissue-specific sample. Five replicates of fat body tissue were created for each species in injected and control groups, and three replicates for hemocyte tissue. RNA was isolated from 3rd instar control or infected larval tissues. For each sample, either fat body from 8 whole larvae or hemocytes from 70 larvae were homogenised in 500 μL Tri-reagent (Ambion 10296010) by pipetting up and down thoroughly with a P1000 pipette. 100 μL of chloroform was added, tubes shaken vigorously for 3 minutes then centrifuged for 10 minutes at 12,000g for 10 minutes. 150 μL of the upper aqueous phase was removed to a fresh tube and 2.5x volumes of propan-2-ol was added and mixed by inverting the tubes several times. After an incubation of 10 minutes at room temperature tubes were centrifuged at 12,000g at 4°C for 10 minutes, the supernatant carefully removed by pipetting and 250 μL ice-cold ethanol was added. The tubes were centrifuged for 2 minutes at 12,000g and 4°C and the ethanol was removed, the RNA pellets were air dried briefly and 30 μL nuclease free water was added and fully dissolved by incubating tubes at 45°C for 5 minutes. RNA was quantified using a Qubit RNA HS assay kit (Thermofisher Q32852). RNAseq libraries were prepared using an Ultra II stranded mRNA library kit (New England Biolabs E7760) with NEB Next multiplex unique indices (New England Biolabs E6440S) and polyA enrichment module (NEB E7490S). Up to 1 μg of total RNA was used to make each library. After polyA enrichment with the polyA enrichment module according to the manufacturer’s recommendation, mRNA was fragmented at 94°C for 8 minutes. Libraries were then prepared according to the manufacturer’s recommendations. Adapter concentrations and number of amplification cycles were adjusted regarding the amount of starting material. Library quantity was measured using Qubit HS DNA assay kit and quality was assessed using a Bioanalyzer high sensitivity kit on an Agilent 2100 Bioanalyzer (Agilent 5067–4626). The libraries were submitted for sequencing at Cancer Research UK Cambridge Institute Genomic Core Facility using Illumina NovaSeq with 150bp single-end reads.

Quantitative PCR

At 24 hours post injection with mineral oil containing wasp homogenate, RNA was extracted from ten groups of 8 whole larvae, along with the same number of extractions from uninfected control larvae for each species (D. simulans, D. sechellia and F1 hybrids). RNA was isolated using RNeasy plus mini columns (Qiagen 74134). The larvae were homogenised in 400 μL of RTL plus buffer supplemented with 4 μL b-mercaptoethanol by crushing with a pestle, then immediately frozen in liquid nitrogen and stored at -70°C. Samples were thawed and centrifuged at maximum speed in a micro-centrifuge for 3 minutes to pellet debris. 350 μL of supernatant was removed to a gDNA eliminator column and RNA was isolated according to the manufacturer’s recommendation, including DNAase treatment. RNA was eluted in 30 μL of nuclease free water.

cDNA was synthesised as follows: for each sample, 1 μL of RNA was added to 0.5 μL of 10 mM of each dNTP, 0.5 μL of 0.5 μg/μL OligodT primer and 4.75 μL of nuclease free water. Samples were incubated at 65°C for 5 minutes and snap frozen on ice for 5 minutes. Next a 4.25 μL master mix containing 2 μL of 5x Superscript IV buffer (Thermofisher #10890010) 0.5 μL DTT, 0.25 μL RNasin plus (Promega N2611) and 0.5 μL Superscript IV. Samples were incubated at 50°C for 10 minutes and then heat inactivated by incubating at 80°C for 10 minutes. cDNA samples were diluted by adding 90 μL of nuclease free water.

The longest conserved constitutive exons (see below) of selected genes from D simulans Sz49 and D sechellia DenisNF100 were used for primer design with Primer-BLAST [58]. qPCR primers were designed so that amplicons contained at least one SNP that differentiated the two species, and the primers contained no SNPs. Each primer pair was tested using a 5x dilution series using hybrid gDNA as a template. Selected primers displayed efficiencies of 100% + /− 5% and single peaks on their melt-curves. qPCR was carried out using SensiFAST Hi-Rox SYBR reagents (Meridian BIO-92020) on a Step-One plus qPCR cycler (Applied Biosystems). Each cDNA was assayed for gene expression using 2 technical replicates in reactions containing 5 μL Sybr, 2 μL nuclease free water, 1 μL of 2.5 μM each primer and 2 μL of diluted cDNA with a fast PCR protocol: 95°C for 2 minutes then 40 amplification cycles of 95°C for 5s, 60°C for 30s followed by a melt curve program consisting of 95°C for 15s then 15s each at 0.3°C increments between 60°C and 95°C. The gene of interest expression was taken from the cycle threshold (Ct), and was normalised with that of the house-keeping gene Rpl32 as ΔCtgene = CtRpl32Ctgene.

Amplicon sequencing

To measure allele-specific expression in specific genes, we created F1 hybrids and amplified the target transcripts using the same primers we used for quantitative PCR. For each hybrid cDNA sample, amplicons from 13 different genes were amplified in 15 μL reactions containing 7.5 μL Q5 High-Fidelity 2X Master Mix (New England Biolabs M0492S), 2 μL of template cDNA, 33 nM each primer and 4 μL of nuclease free water. Primers were designed to incorporate 5’ adapter sequences for the subsequent indexing PCR (see S1 Table for primers). Amplicons were identical to those used for qPCR. To test for PCR biases that favoured the allele of one species over the other species, we included genomic DNA from F1 hybrids as a control. To check that we could assign reads correctly to each species, we included a control of cDNA from the parental D. sechellia and D. simulans lines. A touch-down PCR program was used comprising 95°C for 3 minutes, 10 cycles of 95°C for 30s, 62°C for 15s decreasing by 1°C every cycle, 72°C for 30s, then 25 cycles of 95°C for 30s, 52°C for 15s, 72°C for 30s. Amplicons for all 13 genes were pooled together using equal volumes of PCR product. 30 μL of each pool was purified by using 3x volumes of ProNex beads (Promega NG2001) and processing according to the manufacturer’s recommendations. The purified pools were eluted from the beads by adding 30 μL of elution buffer (10 mM Tris, pH8.0). To add indexes to each pool a further PCR was carried out containing 12.5 μL Q5 High-Fidelity 2X Master Mix, 2.5 μL of Nextera index 1, 2.5 μL of Nextera index 2 (Illumina FC-131–1001), 2.5 μL of purified pool and 5 μL of nuclease free water. The PCR program was 95°C for 3 minutes, 8 cycles of 95°C for 30s, 55°C for 30s, 72°C for 30s, then 72°C for 5 minutes. Indexed libraries were pooled together using equal quantities of PCR products, and 30 μL was taken and purified using 90 μL of ProNex beads as before. The multiplex was eluted from the beads in 30 μL of 2 mM Tris pH8.0 and quantified using Qubit DNA high sensitivity kit (Thermofisher Q32854). The mean fragment size of the multiplex was 220bp, it was diluted to 10 nM with 10 mM Tris pH8.0 and submitted for sequencing at the University of Cambridge, Department of Biochemistry DNA sequencing facility using Illumina MiSeq with 300bp paired end reads.

Digital cytometry

The digital cytometry method CIBERSORTx [32] was used to deconvolute the bulk RNA-seq data and infer hemocyte proportions in the parent Drosophila species and the hybrids using the reference single cell profile generated for D. melanogaster by Leitão et al. [31]. The top 2000 genes that had highly variable expression among cell types in that study and genes that had an average log2 expression greater than or equal to 0.05 across cell types were used to generate a signature matrix profile for the deconvolution. The following default recommended parameters were used for generating the signature matrix profile: a) no quantile normalisation was performed, b) a κ of 999 was used, c) the expression levels of 300–500 genes were used for barcoding cell types, d) a q-value threshold of 0.01 was used to test for differential expression, e) only cell phenotypes that were replicated a minimum of five times were retained and f) half of the available gene expression profiles were randomly sampled to generate the profile. After this, the cell fractions in the bulk RNA-seq were imputed using S-mode batch correction. 100 permutations were used to test the significance of cluster inferences.

Analysis of transcription factor binding sites in fat body immune responsive genes

The FIMO tool [59] in the MEME suite [60] was used for searching the 1-kb upstream regions of coding sequences in D. simulans and D. sechellia genomes for transcription factor binding site (TFBS) motifs. We searched for TFBS motifs of 6 immune related transcription factors (TF): Relish [37, 61], Dif [37, 62, 63], STAT [64], Serpent [65], Dorsal [65] and CrebA [66] (see S7 Dataset for motif sequences). For each TF, the 531 fat body immune responsive genes were categorised as motif present (motif/+) or motif absent (motif/-) in both D. simulans or D. sechellia. The trans-divrged immune responsive genes were also categorised as D. simulans or D. sechellia biased in their divergence according to the log2 fold change estimated from the allele specific expression analysis. For each TF, a χ2 test was carried out to test if trans-divergence was associated with the presence of TFBS motifs (we required the motifs to be present in both D. simulans and D. sechellia) (S2A and S2B Table). Another χ2 test was carried out to test if cis-divergence was associated with genes where TFBS motifs were found in only one of the two species (S2C Table).

Generating alternative genomes for D. simulans and D. sechellia

We used publicly available data to produce reference genomes for the Drosophila lines we were using. Whole genome sequencing data for Sz49 was obtained from NCBI Sequence Read Archive, under BioProject PRJNA318623 (BioSample: SAMN05157406) [54]. Whole genome sequencing data for DenisNF100 was obtained from NCBI Sequence Read Archive, under BioProject PRJNA395473 (BioSample: SAMN07407394) [56].

Genomic sequencing reads of Sz49 and DenisNF100 were mapped to D. simulans reference genome (FlyBase Dsim_r2.01) [67, 68] and the D. sechellia reference genome (FlyBase Dsec_r1.3) [67] respectively using the default parameters of the BWA-MEM algorithm in the BWA package [69]. Duplicated read pairs were removed using Markduplicates in the Picard toolkit (http://broadinstitute.github.io/picard/). Subsequently, variants were called against the reference genome using the GATK (v4.1.4.1) [70] HaplotypeCaller toolkit. Single nucleotide polymorphisms (SNPs) and indels were separated for Base Quality Score Recalibrations (BQSR) [70]. Each round of BQSR (GATK toolkit) was performed using hard-filtered variants from the previous round as “true set”. For SNPs, the hard-filtering criteria were set with QualByDepth < 2.0, StrandOddRatio>3.0, FisherStrand > 60.0, RMSMappingQuality<40.0, MappingQualityRankSumTest < -12.5 and ReadPosRankSumTest < -8.0. For indels, the hard-filtering criteria were set with QualByDepth < 2.0, FisherStrand > 200.0, ReadPosRankSumTest < -20.0. BQSR were performed until the number of output variants plateaued or oscillated around a constant number. The finalised sets of variants after BQSR were then used to modify the reference genomes and generate Sz49 and DenisNF100 genomes with GATK FastaAlternateReferenceMaker toolkit.

Obtaining allele-specific expression estimates

Raw RNA sequencing reads were trimmed using Trim Galore (http://www.bioinformatics.babraham.ac.uk/projects/trim_galore/), setting the low-quality Phred score at 30 for quality control and using the first 13 base pairs from Illumina adaptors for adaptor trimming. Low-quality bases were trimmed from the 3’ end, and reads with fewer than 50 bases remaining were removed.

To remove ambiguity from allele-specific expression data, only the reads that uniquely mapped with no mismatches to the genomes of one species while not mapping to the other species are considered allele-specific reads and included in the subsequent analyses. To make our analysis robust to errors in the Sz49 and DenisNF100 genomes we created, the published reference genomes Dsim_r2.01 and Dsec_r1.3 were also incorporated in the mapping pipeline (S1 Fig). For instance, to obtain RNA sequencing reads from the D. simulans allele, reads were first mapped to our D. sechelliaDenisNF100 genome followed by the Dsec_r1.3 reference genome to identify non-D. sechellia reads. These reads were then collected and mapped to our Sz49 genome, followed by the Dsim_r2.01 reference genome, D. simulans. The reads which mapped to either of these genomes were considered to be from the D. simulans allele. The same procedures were repeated to identify D. sechellia RNA reads for each library. Note that this procedure means that any read that maps to both species is discarded and not included in any of our analyses. As a consequence, our read counts do not reflect absolute expression levels of a gene. Read mapping was performed using bowtie [71] with settings allowing 0 mismatches and up to 3 reportable alignments. Only the reads that had a single reported alignment were retained for analysis.

Expression was represented by the number of reads mapped to the gene in question. To ensure we are studying differences in expression level rather than patterns of alternative splicing, we only considered exons that were present in all the transcripts produced by a gene (‘constitutive exons’). To identify the location of homologous exons in the two species, we needed to convert the genome coordinates of an exon in one species to the genome coordinates of the other species. Furthermore, to prevent changes in gene length being mistaken for changes in expression, we only included exon sequence that was conserved between D. simulans and D. sechellia (‘conserved exons’). Exons only present in one species were not considered, and where the exon boundaries had changed, we altered the exon boundaries so that they were the same in both species. The approach we took was to convert the genomic coordinates of each read mapped to the constitutive exons of one species to the coordinates of the other species, and we discarded the read if it did not overlap with an exon in that species. This genomic coordinates conversion was done using the LiftOver tool developed by UCSC [72]. The chain files for converting Dsim_r2.01 and Dsec_r1.3 coordinates were provided by UCSC Genomic Institute. The chain files for Sz49 to Dsim_r2.01 and DenisNF100 to Dsec_r1.3 coordinates conversion were generated locally with UCSC command line tools. Read counts were generated with HTSeq-count [73]. The procedures of annotating allele-specific gene expressions are detailed in S3 Fig. This pipeline is designed to accommodate the available chain files and published annotation files for Dsim_r2.01 and Dsec_r1.3.

Statistical analyses of RNAseq data

Low expression genes that had a count per million (CPM) < = 3 in at least 3 hemocytes libraries or at least 5 fat body libraries were filtered out. A CPM of 3 is equivalent to 10 mapped reads in the smallest hemocytes library and 18 mapped reads in the smallest fat body library. The R package edgeR [74] was used for differential expression analyses with RNAseq data. Dispersions were measured using the Cox-Reid profile adjusted likelihood method before fitting the expression data with a negative binomial generalised linear model. Differential expression was determined using a quasi-likelihood F-test, and a false discovery rate (FDR) of 0.05 was set as the significance threshold.

To compare expression levels, we used a generalized linear model (GLM) implemented in edgeR [74]. This allows us to perform different contrasts both to estimate P values and fold changes in gene expression. First, to examine the expression patterns when flies mount an immune response, we analysed each species and F1 hybrids separately, and the contrast was set between the immune challenged and control samples. Second, to identify genes that were deferentially expressed between D. simulans and D. sechellia, we analysed the immune challenged and control groups separately, and the contrast was set between species. Third, we examined how the immune response had diverged between species. To achieve this, we contrasted the transcriptional response to immune challenge in D. simulans to the transcriptional response in D. sechellia.

These comparisons between species detect change in gene expression caused by both (cis and trans factors. To investigate change in cis alone, we looked for differences in the expression of the two alleles in F1 hybrids. To detect allele-specific differential expression, we repeated the contrasts between species, but this time the data was read counts from the two alleles in the F1 hybrids.

Having analysed divergence between the species (cis and trans) and allele-specific expression (cis), we could then use the difference between these to estimate divergence in trans. Again, the magnitude and significance of trans effects was estimated by performing this contrast within the GLM framework of edgeR.

We placed genes within different categories of expression divergence following McManus, 2010 [14]. Cis: significant ASE within F1 hybrids, no significant trans divergence; Trans: significant trans divergence but no significant ASE within F1 hybrids; Cis + Trans: ASE within F1 hybrids, divergence between parents and divergence in trans are all significant, with cis and trans divergence in the same direction; Cis x Trans: ASE within F1 hybrids, divergence between parents and divergence in trans are all significant, with cis and trans divergence in the opposite direction; Compensatory: significant ASE within F1 hybrids and in trans, but no significant divergence between parents; Conserved: no significant change in gene expression in any of the comparisons; Ambiguous: all the rest. We noticed that many ambiguous genes resulted from our arbitrary choice of significance threshold – the divergence between species crossed the threshold, but cis and trans effects did not. Among the ambiguous genes, we therefore examined the magnitude of the cis and trans log fold change estimates. If the cis or trans effect explained at least half the divergence of the two species, we classified the gene as ‘cis’ or ‘trans’. Cis diverged genes include those categorised as Cis, Cis + Trans, Cis x Trans and Compensatory. Trans diverged genes include those categorised as Trans, Cis + Trans, Cis x Trans and Compensatory. Both Cis and Trans diverged genes include those categorised as Cis + Trans, Cis x Trans and Compensatory.

Gene ontology enrichment analysis was performed using FlyMine [75] database and tools, with the 4969 analysed fat boy genes as background population.

Accuracy of estimates of allele-specific expression

To investigate the relative contribution of cis- and trans- regulatory changes to the expression divergence between D. simulans and D. sechellia, we compared the expression of the two alleles in F1 hybrids (allele-specific expression). To do this, we assigned RNAseq reads to D. simulans and D. sechellia using sequence differences between the species (S1 and S3 Figs). As the two alleles share the same trans-regulatory environment in hybrids, allele-specific expression is a measure of cis- regulatory divergence between the species [26]. Expression differences between the parental species is caused by both cis and trans divergence, so trans effects can be estimated by subtracting the cis effects from the total divergence between parents [10].

We used two approaches to confirm we could accurately assign RNAseq reads to the correct species. First, because all the data went through the same analysis pipelines (S1 and S3 Figs), we can estimate our error rate by checking whether reads from the parental species were always assigned to that species. This allowed us to filter out genes with a > 0.01 mismapping rate in either parental species. After also removing genes with low expression levels, there were 4969 and 5275 genes remaining in fat body and hemocyte data-sets respectively. In this dataset, we could successfully allocate the large majority of reads to the correct species, although more mismapping happened to reads from D. simulans (S2(A) Fig), likely due to higher genetic diversity in this species. Two hemocytes libraries (B7 and D7) had a low level of contamination between species (S2(A) and S2(C) Fig). Secondly, we compared the total number of reads from the two species in F1 hybrids. In these libraries, read counts from D. simulans and D. sechellia are very similar, again suggesting no systematic bias in assigning reads to species (S2(B) and S2(C) Fig).

Quantifying allele-specific expression using amplicon sequencing

Following amplicon sequencing, Illumina MiSeq reads of the 13 selected genes were mapped to the D. simulans (Sz49) genome sequence using the default parameters of the BWA-MEM algorithm in the BWA package [69]. The amplicons had been selected so that they contained SNPs that allowed us to distinguish reads from the D. simulans and D. sechellia alleles. We checked that these were homozygous in the parental species by visual inspection in Integrative Genomics Viewer (IGV_2.9.4) [76].

To estimate the relative number of transcripts from the D. simulans and D. sechellia alleles in F1 hybrids, we counted the number of reads with each SNP allele using tools implemented in Rsamtools [77]. For genes with more than one SNP in the amplicon, we confirmed that the different SNPs resulted in similar estimates of allele-specific expression. As this was the case, we only analysed data from the first SNP from the 5’ end. We tested if there was a bias towards the D. simulans or D. sechellia alleles by analysing reads generated by amplifying genomic DNA from F1 hybrids, and checking half the reads came from each species.

Transcript counts for each gene were analysed by fitting a mixed effects logistic regression using the glmer function in the R package lme4 [78]. We treated allele (D. simulans/D. sechellia) and treatment (control/infected) as fixed effects and sample identity as a random effect (i.e., this random effect estimated the residual variance and therefore accounts for over-dispersion). As a measure of the extent of cis divergence, we estimated the log odds (logit) of an RNAseq read being derived from the D. simulans allele under either controlled or infected conditions. This parameter was extracted from estimated coefficients in the regression model. A logit equal to zero suggest no difference between the expression of the two species’ alleles in hybrids, and therefore no interspecific cis-regulatory divergence.

Statistical analysis of qPCR data

Expression levels of the 13 selected genes were obtained from qPCR cycle thresholds (CT) normalised using the reference gene Rpl32CT). In the F1 hybrids, we then multiplied these ΔCT values by the proportion of transcripts from the D. simulans and D. sechellia alleles (allele-specific expression, estimated as described in the previous section), giving us the expression of each allele separately. The ΔCT values in the parental species were divided by two to make them comparable to the expression levels in hybrids.

To estimate trans-regulatory divergence in these genes, we assessed the goodness of fit of a full model fitted with relative expression levels against that of a series of its nested models using likelihood ratio test implemented by R package lmtest [79]. For the full model, the allelic expression levels of each gene (CT values from PCR programs) from both parental and hybrid libraries were fitted into a general linear regression with ‘lm’ function in R package stats [80], with allelic type (D. simulans/D. sechellia), species (parents/hybrids), treatment (controlled/infected) and their interactions (allelic type:species, allelic type:treatment, species:treatment, allelic type:species:treatment) as parameters. We compared the full model with all 7 parameters with the nested model with a subset of parameters (allelic type, treatment, allelic type:treatment) excluding terms concerning “species” to evaluate the interspecific divergence of trans-effect on relative gene expressions, i.e., differential expression patterns between parental alleles and hybrid alleles. The trans contribution to interspecific divergence under controlled or infected conditions was estimated from the coefficient measuring “allele type” and “species” interaction (species:allele type) in the full regression model, i.e., the relative abundance between allele transcripts depending on whether in parental species or hybrids. The controlled and infected trans divergences were inferred from the full model by setting the intercept at “control” and “wasp” status, respectively. The effect of trans-regulatory divergence on immune response was represented by the three-way interaction term of the parameters (allele type:species:treatment) in the full regression model. It is the difference of trans contribution in interspecies divergence between control and infected status.

Supporting information

S1 Dataset. RNAseq read counts for immune responsive genes in hemocytes.

The column titles include the type of sample in upper case (D. sechellia, D. simulans or hybrid), and the allele the reads were assigned to in lower case (D. sechellia or D. simulans).

https://doi.org/10.1371/journal.pgen.1010453.s001

(CSV)

S2 Dataset. RNAseq read counts for immune responsive genes in the fat body.

The column titles include the type of sample in upper case (D. sechellia, D. simulans or hybrid), and the allele the reads were assigned to in lower case (D. sechellia or D. simulans).

https://doi.org/10.1371/journal.pgen.1010453.s002

(CSV)

S3 Dataset. Differential expression of genes in response to immune challenge within each species.

This data was used to plot Figs 2A, 2B and 3B.

https://doi.org/10.1371/journal.pgen.1010453.s003

(CSV)

S4 Dataset. Proportion of lamellocytes and their precursors inferred using digital cytometry.

This data was used to plot Fig 3A.

https://doi.org/10.1371/journal.pgen.1010453.s004

(CSV)

S5 Dataset. Differential expression of genes in cis and trans in hemocytes.

This data was used to plot Fig 4.

https://doi.org/10.1371/journal.pgen.1010453.s005

(CSV)

S6 Dataset. Differential expression of genes in cis and trans in hemocytes.

This data was used to plot Fig 5.

https://doi.org/10.1371/journal.pgen.1010453.s006

(CSV)

S7 Dataset. Transcription factor binding motifs.

https://doi.org/10.1371/journal.pgen.1010453.s007

(TXT)

S8 Dataset. Read counts from amplicon sequencing of selected immunity genes.

This data underlies Figs 6 and 7.

https://doi.org/10.1371/journal.pgen.1010453.s008

(CSV)

S9 Dataset. Gene expression of selected genes estimated by quantitative PCR.

The data is raw cycle threshold values, and was combined with S8 Dataset to generate Fig 7.

https://doi.org/10.1371/journal.pgen.1010453.s009

(XLS)

S10 Dataset. Read counts per million (cpm) for immune responsive genes in the fat body.

Separate cpm values are given for each library. This data underlies Fig 3B, and was combined with the statistical significance values in S3 Dataset to generate Fig 3B.

https://doi.org/10.1371/journal.pgen.1010453.s010

(CSV)

S11 Dataset. Differential expression of genes between D. simulans and D. sechellia.

This data was used to generate Fig 5.

https://doi.org/10.1371/journal.pgen.1010453.s011

(CSV)

S1 Table. Primer sequences used for both quantitative PCR and amplicon sequencing.

https://doi.org/10.1371/journal.pgen.1010453.s012

(PDF)

S2 Table. Number of cis or trans diverged immune responsive genes in the fat body, with or without transcription factor binding site motifs.

Immune-responsive genes in the fat body were classified as to whether they had motifs associated with the binding of six transcription factors. In (A) and (B) Motif/+ genes had the motif in both D. sechellia and D. simulans. In (C) Motif+ genes had the motif in just one of these species but not the other. These genes in turn were classified as to whether they had diverged in cis or trans in control or immune-challenged conditions, of if the response to infection had been changed in trans. The P value is from a chi-squared test.

https://doi.org/10.1371/journal.pgen.1010453.s013

(PDF)

S1 Fig. Bioinformatic pipeline to obtain allele-specific RNA sequencing read counts.

https://doi.org/10.1371/journal.pgen.1010453.s014

(PDF)

S2 Fig. Accuracy of allele-specific RNA-seq read mapping.

(A) The proportion of reads from single-species libraries that were incorrectly mapped to the wrong species. (B) The proportion of reads that were mapped to different species’ genomes. Each point is a different sequencing library. The colours reflect the species from which that library was made.

https://doi.org/10.1371/journal.pgen.1010453.s015

(PDF)

S3 Fig. Obtaining allele-specific gene expressions for each library.

In each library, aligned coordinates of mapped to Sz49 genome reads were converted to Dsim_r2.01 coordinates then Dsec_r1.3 coordinates with LiftOver, allowing selection for mapped to Dsec_r1.3 constitutive exons reads. The selected reads, now with Dsec_r1.3 coordinates, would be converted to Dsim_r2.01 coordinates, allowing us to see whether the read mapping to exon sequence that is found in both species (‘conserved exons’). This allowed us to use HTSeq-count to produce the read counts just those reads mapping to exon sequence present in both species. Similarly, reads mapped to Dsim_r1.2, DsecNF100 and Dsec_r1.3 genomes went through various coordinate conversions, so we could check they mapped to sequence annotated as an exon in both species’ reference genomes (Dsec_r1.3 and Dsim_r2.01). Any read that mapped to sequence annotated as an exon in only one of the reference genomes was discarded.

https://doi.org/10.1371/journal.pgen.1010453.s016

(PDF)

S4 Fig. Dominance of gene expression in F1 hybrids.

Differences in gene expression between the parental species and hybrids in hemocytes and fat body. (A) The scatter plots comparing the relative expression differences between F1 hybrids and each parental species. Each point is an immune-responsive gene (a gene that was differentially expressed after immune challenge in one or more species). The colours represent different dominance relationships. Under- and over-dominant genes have lower or higher expression in hybrids than both parental species. Additive genes have an expression level in hybrids that is intermediate between the parents. Dominant genes have expression levels that resemble one parent. Following McManus 2010 [14] and Gibson 2004 [81], expression was considered different between hybrids and the parents if it was both statistically significant (FDR<0.05) and there was at least 1.25 log2 fold difference. (B) The number of the genes in each category.

https://doi.org/10.1371/journal.pgen.1010453.s017

(PDF)

S5 Fig. Proportion of mature and immature lamellocytes among hemocytes.

Proportions of lamellocytes on different transcriptional states in D. simulans, D. sechellia and F1 hybrid larvae under control and immune challenged conditions, estimated from bulk RNA-seq data with CIBERSORTx. The upper and lower lines on top of the bar give the 95% confidence interval of the estimate.

https://doi.org/10.1371/journal.pgen.1010453.s018

(PDF)

Acknowledgments

We thank Daniel Matute and Sarah Signor for providing the D. sechellia and D. simulans flies.

References

  1. 1. Schlenke TA, Morales J, Govind S, Clark AG. Contrasting infection strategies in generalist and specialist wasp parasitoids of Drosophila melanogaster. PLoS pathogens. 2007;3(10):e158. pmid:17967061
  2. 2. Leitão AB, Arunkumar R, Day JP, Hanna N, Devi A, Hayes MP, et al. Recognition of non-self is necessary to activate Drosophila’s immune response against an insect parasite. bioRxiv. 2022;.
  3. 3. Márkus R, Kurucz É, Rus F, Andó I. Sterile wounding is a minimal and sufficient trigger for a cellular immune response in Drosophila melanogaster. Immunology letters. 2005;101(1):108–111. pmid:15964636
  4. 4. Lemaitre B, Hoffmann J. The host defense of Drosophila melanogaster. Annu Rev Immunol. 2007;25:697–743. pmid:17201680
  5. 5. De Gregorio E, Spellman PT, Rubin GM, Lemaitre B. Genome-wide analysis of the Drosophila immune response by using oligonucleotide microarrays. Proceedings of the National Academy of Sciences. 2001;98(22):12590–12595. pmid:11606746
  6. 6. Fauvarque MO, Williams MJ. Drosophila cellular immunity: a story of migration and adhesion. Journal of cell science. 2011;124(9):1373–1382. pmid:21502134
  7. 7. Sackton TB, Lazzaro BP, Schlenke TA, Evans JD, Hultmark D, Clark AG. Dynamic evolution of the innate immune system in Drosophila. Nature genetics. 2007;39(12):1461–1468. pmid:17987029
  8. 8. Obbard DJ, Welch JJ, Kim KW, Jiggins FM. Quantifying adaptive evolution in the Drosophila immune system. PLoS genetics. 2009;5(10):e1000698. pmid:19851448
  9. 9. Unckless RL, Howick VM, Lazzaro BP. Convergent balancing selection on an antimicrobial peptide in Drosophila. Current Biology. 2016;26(2):257–262. pmid:26776733
  10. 10. Wittkopp PJ, Haerum BK, Clark AG. Evolutionary changes in cis and trans gene regulation. Nature. 2004;430(6995):85–88. pmid:15229602
  11. 11. Wittkopp P, Haerum B, Clark A. Genetic basis of regulatory variation within and between Drosophila species. Nat Genet. 2008;40:346–350.
  12. 12. Wilson MD, Barbosa-Morais NL, Schmidt D, Conboy CM, Vanes L, Tybulewicz VL, et al. Species-specific transcription in mice carrying human chromosome 21. Science. 2008;322(5900):434–438. pmid:18787134
  13. 13. Tirosh I, Reikhav S, Levy AA, Barkai N. A yeast hybrid provides insight into the evolution of gene expression regulation. Science. 2009;324(5927):659–662. pmid:19407207
  14. 14. McManus CJ, Coolon JD, Duff MO, Eipper-Mains J, Graveley BR, Wittkopp PJ. Regulatory divergence in Drosophila revealed by mRNA-seq. Genome research. 2010;20(6):816–825. pmid:20354124
  15. 15. Signor SA, Nuzhdin SV. The evolution of gene expression in cis and trans. Trends in Genetics. 2018;34(7):532–544. pmid:29680748
  16. 16. Snoek BL, Sterken MG, Bevers RP, Volkers RJ, van’t Hof A, Brenchley R, et al. Contribution of trans regulatory eQTL to cryptic genetic variation in C. elegans. BMC genomics. 2017;18(1):1–15. pmid:28662696
  17. 17. Lazzaro BP, Sceurman BK, Clark AG. Genetic basis of natural variation in D. melanogaster antibacterial immunity. Science. 2004;303(5665):1873–1876. pmid:15031506
  18. 18. Ramirez-Corona BA, Fruth S, Ofoegbu O, Wunderlich Z. The mode of expression divergence in Drosophila fat body is infection-specific. Genome research. 2021;31(6):1024–1034. pmid:33858842
  19. 19. Rizki TM, Rizki RM. Lamellocyte differentiation in Drosophila larvae parasitized by Leptopilina. Developmental & Comparative Immunology. 1992;16(2-3):103–110. pmid:1499832
  20. 20. Nappi A, Poirie M, Carton Y. The role of melanization and cytotoxic by-products in the cellular immune responses of Drosophila against parasitic wasps. Advances in parasitology. 2009;70:99–121. pmid:19773068
  21. 21. Salazar-Jaramillo L, Wertheim B. Does Drosophila sechellia escape parasitoid attack by feeding on a toxic resource? PeerJ. 2021;9:e10528. pmid:33505786
  22. 22. Salazar-Jaramillo L, Jalvingh KM, de Haan A, Kraaijeveld K, Buermans H, Wertheim B. Inter-and intra-species variation in genome-wide gene expression of Drosophila in response to parasitoid wasp attack. BMC genomics. 2017;18(1):1–14. pmid:28449654
  23. 23. Salazar-Jaramillo L, Paspati A, van de Zande L, Vermeulen CJ, Schwander T, Wertheim B. Evolution of a cellular immune response in Drosophila: a phenotypic and genomic comparative analysis. Genome biology and evolution. 2014;6(2):273–289. pmid:24443439
  24. 24. Eslin P, Prévost G. Hemocyte load and immune resistance to Asobara tabida are correlated in species of the Drosophila melanogaster subgroup. Journal of Insect Physiology. 1998;44(9):807–816. pmid:12769876
  25. 25. Obbard DJ, Maclennan J, Kim KW, Rambaut A, O’Grady PM, Jiggins FM. Estimating divergence dates and substitution rates in the Drosophila phylogeny. Molecular biology and evolution. 2012;29(11):3459–3473. pmid:22683811
  26. 26. Cowles CR, Hirschhorn JN, Altshuler D, Lander ES. Detection of regulatory variation in mouse genes. Nature genetics. 2002;32(3):432–437. pmid:12410233
  27. 27. Yan H, Yuan W, Velculescu VE, Vogelstein B, Kinzler KW. Allelic variation in human gene expression. Science. 2002;297(5584):1143–1143. pmid:12183620
  28. 28. Kim-Jo C, Gatti JL, Poirié M. Drosophila cellular immunity against parasitoid wasps: A complex and time-dependent process. Frontiers in Physiology. 2019;10:603. pmid:31156469
  29. 29. Wertheim B, Kraaijeveld AR, Schuster E, Blanc E, Hopkins M, Pletcher SD, et al. Genome-wide gene expression in response to parasitoid attack in Drosophila. Genome biology. 2005;6(11):1–20. pmid:16277749
  30. 30. Irving P, Ubeda JM, Doucet D, Troxler L, Lagueux M, Zachary D, et al. New insights into Drosophila larval haemocyte functions through genome-wide analysis. Cellular microbiology. 2005;7(3):335–350. pmid:15679837
  31. 31. Leitão AB, Arunkumar R, Day JP, Geldman EM, Morin-Poulard I, Crozatier M, et al. Constitutive activation of cellular immunity underlies the evolution of resistance to infection in Drosophila. Elife. 2020;9:e59095. pmid:33357377
  32. 32. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nature biotechnology. 2019;37(7):773–782. pmid:31061481
  33. 33. Lindsay SA, Lin SJ, Wasserman SA. Short-Form bomanins mediate humoral immunity in Drosophila. Journal of innate immunity. 2018;10(4):306–314. pmid:29920489
  34. 34. Clemmons AW, Lindsay SA, Wasserman SA. An effector peptide family required for Drosophila Toll-mediated immunity. PLoS Pathogens. 2015;11(4):e1004876. pmid:25915418
  35. 35. Ekengren S, Hultmark D. A family of Turandot-related genes in the humoral stress response of Drosophila. Biochemical and biophysical research communications. 2001;284(4):998–1003. pmid:11409894
  36. 36. Kurucz É, Váczi B, Márkus R, Laurinyecz B, Vilmos P, Zsámboki J, et al. Definition of Drosophila hemocyte subsets by cell-type specific antigens. Acta Biologica Hungarica. 2007;58(Supplement-1):95–111. pmid:18297797
  37. 37. Hanson MA, Cohen LB, Marra A, Iatsenko I, Wasserman SA, Lemaitre B. The Drosophila Baramicin polypeptide gene protects against fungal infection. PLoS pathogens. 2021;17(8):e1009846. pmid:34432851
  38. 38. Clemmons AW, Lindsay SA, Wasserman SA. An effector peptide family required for Drosophila Toll-mediated immunity. PLoS Pathogens. 2015;11(4):e1004876. pmid:25915418
  39. 39. Tanji T, Hu X, Weber AN, Ip YT. Toll and IMD pathways synergistically activate an innate immune response in Drosophila melanogaster. Molecular and cellular biology. 2007;27(12):4578–4588. pmid:17438142
  40. 40. Vanha-Aho LM, Anderl I, Vesala L, Hultmark D, Valanne S, Rämet M. Edin expression in the fat body is required in the defense against parasitic wasps in Drosophila melanogaster. PLoS pathogens. 2015;11(5):e1004895. pmid:25965263
  41. 41. Prud’homme B, Gompel N, Carroll SB. Emerging principles of regulatory evolution. Proceedings of the National Academy of Sciences. 2007;104(suppl 1):8605–8612. pmid:17494759
  42. 42. Stern DL, Orgogozo V. The loci of evolution: how predictable is genetic evolution? Evolution: International Journal of Organic Evolution. 2008;62(9):2155–2177. pmid:18616572
  43. 43. Wittkopp P. Genomic sources of regulatory variation in cis and in trans. Cellular and Molecular Life Sciences CMLS. 2005;62(16):1779–1783. pmid:15968467
  44. 44. Strand MR, Pech LL. Immunological basis for compatibility in parasitoid-host relationships. Annual review of entomology. 1995;40(1):31–56. pmid:7810989
  45. 45. Benassi V, Coustau C, Carton Y. Insect immunity: a genetic factor (hrtp) is essential for antibacterial peptide expression in Drosophila after infection by parasitoid wasps. Archives of Insect Biochemistry and Physiology: Published in Collaboration with the Entomological Society of America. 2000;43(2):64–71. pmid:10644970
  46. 46. Boutros M, Agaisse H, Perrimon N. Sequential activation of signaling pathways during innate immune responses in Drosophila. Developmental cell. 2002;3(5):711–722. pmid:12431377
  47. 47. Dudzic JP, Hanson MA, Iatsenko I, Kondo S, Lemaitre B. More than black or white: melanization and toll share regulatory serine proteases in Drosophila. Cell reports. 2019;27(4):1050–1061. pmid:31018123
  48. 48. Lavine M, Strand M. Insect hemocytes and their role in immunity. Insect biochemistry and molecular biology. 2002;32(10):1295–1309. pmid:12225920
  49. 49. Carton Y, Nappi A. Drosophila cellular immunity against parasitoids. Parasitology today. 1997;13(6):218–227. pmid:15275074
  50. 50. Meister M. Blood cells of Drosophila: cell lineages and role in host defence. Current opinion in immunology. 2004;16(1):10–15. pmid:14734104
  51. 51. Brennan CA, Delaney JR, Schneider DS, Anderson KV. Psidin is required in Drosophila blood cells for both phagocytic degradation and immune activation of the fat body. Current Biology. 2007;17(1):67–72. pmid:17208189
  52. 52. Shia AK, Glittenberg M, Thompson G, Weber AN, Reichhart JM, Ligoxygakis P. Toll-dependent antimicrobial responses in Drosophila larval fat body require Spatzle secreted by haemocytes. Journal of cell science. 2009;122(24):4505–4515. pmid:19934223
  53. 53. Schmid MR, Anderl I, Vesala L, Vanha-aho LM, Deng XJ, Rämet M, et al. Control of Drosophila blood cell activation via Toll signaling in the fat body. PloS one. 2014;9(8):e102568. pmid:25102059
  54. 54. Signor SA, New FN, Nuzhdin S. A large panel of Drosophila simulans reveals an abundance of common variants. Genome biology and evolution. 2018;10(1):189–206. pmid:29228179
  55. 55. Matute DR, Ayroles JF. Hybridization occurs between Drosophila simulans and D. sechellia in the Seychelles archipelago. Journal of evolutionary biology. 2014;27(6):1057–1068. pmid:24773151
  56. 56. Schrider DR, Ayroles J, Matute DR, Kern AD. Supervised machine learning reveals introgressed loci in the genomes of Drosophila simulans and D. sechellia. PLoS genetics. 2018;14(4):e1007341. pmid:29684059
  57. 57. Longdon B, Hadfield JD, Day JP, Smith SC, McGonigle JE, Cogni R, et al. The causes and consequences of changes in virulence following pathogen host shifts. PLoS pathogens. 2015;11(3):e1004728. pmid:25774803
  58. 58. Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S, Madden TL. Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC bioinformatics. 2012;13(1):1–11. pmid:22708584
  59. 59. Grant CE, Bailey TL, Noble WS. FIMO: scanning for occurrences of a given motif. Bioinformatics. 2011;27(7):1017–1018. pmid:21330290
  60. 60. Bailey TL, Boden M, Buske FA, Frith M, Grant CE, Clementi L, et al. MEME SUITE: tools for motif discovery and searching. Nucleic acids research. 2009;37(suppl_2):W202–W208. pmid:19458158
  61. 61. Copley RR, Totrov M, Linnell J, Field S, Ragoussis J, Udalova IA. Functional conservation of Rel binding sites in drosophilid genomes. Genome research. 2007;17(9):1327–1335. pmid:17785540
  62. 62. Busse MS, Arnold CP, Towb P, Katrivesis J, Wasserman SA. A κB sequence code for pathway-specific innate immune responses. The EMBO journal. 2007;26(16):3826–3835. pmid:17660749
  63. 63. Tanji T, Yun EY, Ip YT. Heterodimers of NF-κB transcription factors DIF and Relish regulate antimicrobial peptide genes in Drosophila. Proceedings of the National Academy of Sciences. 2010;107(33):14715–14720. pmid:20679214
  64. 64. Betz A, Ryoo HD, Steller H, Darnell JE Jr. STAT92E is a positive regulator of Drosophila inhibitor of apoptosis 1 (DIAP/1) and protects against radiation-induced apoptosis. Proceedings of the National Academy of Sciences. 2008;105(37):13805–13810. pmid:18779571
  65. 65. Senger K, Armstrong GW, Rowell WJ, Kwan JM, Markstein M, Levine M. Immunity regulatory DNAs share common organizational features in Drosophila. Molecular cell. 2004;13(1):19–32. pmid:14731391
  66. 66. Shazman S, Lee H, Socol Y, Mann RS, Honig B. OnTheFly: a database of Drosophila melanogaster transcription factors and their binding sites. Nucleic acids research. 2014;42(D1):D167–D171. pmid:24271386
  67. 67. Clark AG, Eisen MB, Smith DR, Bergman CM, Oliver B, Markow TA, et al. Evolution of genes and genomes on the Drosophila phylogeny. Nature. 2007;450(7167):203–218. pmid:17994087
  68. 68. Hu TT, Eisen MB, Thornton KR, Andolfatto P. A second-generation assembly of the Drosophila simulans genome provides new insights into patterns of lineage-specific divergence. Genome research. 2013;23(1):89–98. pmid:22936249
  69. 69. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv preprint arXiv:13033997. 2013;.
  70. 70. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome research. 2010;20(9):1297–1303. pmid:20644199
  71. 71. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome biology. 2009;10(3):1–10. pmid:19261174
  72. 72. Haeussler M, Zweig AS, Tyner C, Speir ML, Rosenbloom KR, Raney BJ, et al. The UCSC genome browser database: 2019 update. Nucleic acids research. 2019;47(D1):D853–D858. pmid:30407534
  73. 73. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. bioinformatics. 2015;31(2):166–169. pmid:25260700
  74. 74. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. pmid:19910308
  75. 75. Lyne R, Smith R, Rutherford K, Wakeling M, Varley A, Guillier F, et al. FlyMine: an integrated database for Drosophila and Anopheles genomics. Genome biology. 2007;8(7):1–16. pmid:17615057
  76. 76. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nature biotechnology. 2011;29(1):24–26. pmid:21221095
  77. 77. Morgan M, Pagès H, Obenchain V, Hayden N. Rsamtools: Binary alignment (BAM), FASTA, variant call (BCF), and tabix file import. R package version. 2016;1(0):677–689.
  78. 78. Bates D, Mächler M, Bolker B, Walker S. Fitting linear mixed-effects models using lme4. arXiv preprint arXiv:14065823. 2014;.
  79. 79. Zeileis A, Hothorn T, et al. Diagnostic checking in regression relationships. 2002;.
  80. 80. Team RC, et al. R: A language and environment for statistical computing. 2013;.
  81. 81. Gibson G, Riley-Berger R, Harshman L, Kopp A, Vacha S, Nuzhdin S, et al. Extensive sex-specific nonadditivity of gene expression in Drosophila melanogaster. Genetics. 2004;167(4):1791–1799. pmid:15342517